WO2009015037A2 - 5-pyridinone substituted indazoles - Google Patents

5-pyridinone substituted indazoles Download PDF

Info

Publication number
WO2009015037A2
WO2009015037A2 PCT/US2008/070535 US2008070535W WO2009015037A2 WO 2009015037 A2 WO2009015037 A2 WO 2009015037A2 US 2008070535 W US2008070535 W US 2008070535W WO 2009015037 A2 WO2009015037 A2 WO 2009015037A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound according
mmol
pyridin
formula
phenyl
Prior art date
Application number
PCT/US2008/070535
Other languages
French (fr)
Other versions
WO2009015037A3 (en
Inventor
Peter Guzzo
Matthew David Surman
Alan John Henderson
Mark Hadden
Original Assignee
Albany Molecular Research, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CA2693377A priority Critical patent/CA2693377A1/en
Priority to CN200880107571A priority patent/CN101861311A/en
Priority to NZ582586A priority patent/NZ582586A/en
Priority to AT08796320T priority patent/ATE544759T1/en
Priority to BRPI0814772-8A2A priority patent/BRPI0814772A2/en
Priority to JP2010518311A priority patent/JP2010534248A/en
Application filed by Albany Molecular Research, Inc. filed Critical Albany Molecular Research, Inc.
Priority to AU2008279321A priority patent/AU2008279321B2/en
Priority to EP08796320A priority patent/EP2176251B1/en
Priority to ES08796320T priority patent/ES2382982T3/en
Publication of WO2009015037A2 publication Critical patent/WO2009015037A2/en
Publication of WO2009015037A3 publication Critical patent/WO2009015037A3/en
Priority to HK10109173.8A priority patent/HK1142608A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/08Bridged systems

Definitions

  • the invention relates to human melanin- concentrating hormone (MCHi) receptor- selective antagonist 5-pyridinone substituted indazoles that are useful for treating obesity, to pharmaceutical compositions comprising these compounds, and to methods for the treatment of obesity, anxiety, depression, and psychiatric disorders in a mammal.
  • MHi human melanin- concentrating hormone
  • MCH Melanin-concentrating hormone
  • mice lacking MCH are hypophagic and lean with increased metabolic rate, whereas animals over-expressing MCH gain excess weight on both standard and high fat diets. MCH is thought to have effects on other nervous system functions as well (Rocksz, L. L. Biological Examination of Melanin Concentrating Hormone 1 : Multi-tasking from the hypothalamus Drug News Perspect 19(5), 273, 2006).
  • GPCR G-protein coupled receptor
  • MCH antagonism was recently identified as a receptor for MCH. Disruption of the binding between MCH and the MCH receptor, i.e. MCH antagonism, may thus be used to counteract the effects of MCH (McBriar, M. D. Recent advances in the discovery of melanin-concentrating hormone receptor antagonists Curr. Opin. Drug Disc. & Dev. 9(4), 496, 2006).
  • R is NR 1 R 2 , wherein R 1 and R 2 are each independently H and optionally substituted alkyl, or R 1 and R 2 , together with the N atom to which they are attached, form a 4-7 membered optionally substituted non-aromatic heterocyclic ring which optionally contains 1 or 2 heteroatoms in addition to the N atom shown; R 3 and R 4 are each independently H or alkyl; or R may combine with either R 3 or R 4 to form an optionally substituted pyrrolidin-2-yl; B is selected from ary, heteroaryl, and cycloalkyl; R 5 , R 6 , R 7 are each independently selected from H, -OH, -O-alkyl, alkyl, halo, -CF 3 , and -CN, -O-aryl, heteroaryl, and hetercyclyl; and R 14 is H or -OH; and provided that when n is 0, R 3 and R 4 are H and R
  • n when n is 0, R 3 and R 4 are H and R is pyrrolidin-1-yl, then when B is a bicyclic aromatic heterocycle, either (i) at least one heteroatom of said bicyclic aromatic heterocycle is adjacent to the position where B attaches to the pyridinone moiety, or (ii) the ring of said bicyclic aromatic heterocycle which attaches to the pyridinone moiety does not contain a heteroatom.
  • R is selected from the group consisting of pyrrolidin-1-yl, 3-hydroxypyrrolidin-l-yl, morpholin-4-yl, 3- hydroxymethylpyrrolidin-1-yl, dimethylamino, piperazin-1-yl, amino, and 2-oxa-5- azabicyclo[2.2.1]heptane-5-yl.
  • R is selected from S-3- hydroxypyrrolidin- 1 -yl, R-3 -hydroxypyrrolidin- 1 -yl, S-3 -hydroxymethylpyrro lidin- 1 -yl, R- 3-hydroxymethylpyrrolidin-l-yl, and (liS ⁇ iS ⁇ -oxa-S-azabicycloP ⁇ .lJheptane-S-yl.
  • R combines with either R 3 or R 4 to form pyrrolidin-2-yl.
  • R 3 and R 4 are both H.
  • n is 0. In other embodiments, n is 1.
  • B is phenyl.
  • B, taken together with R 5 , R 6 and R 7 is selected from the group consisting of phenyl, 4-chlorophenyl, 4-fluorophenyl, 3-chlorophenyl, 3-phenoxyphenyl, 4-trifluoromethylphenyl, 3- trifluoromethylphenyl, 2,4-dichlorophenyl, 4-methylphenyl, 4-trifluoromethoxyphenyl, A- cyanophenyl, 4-methoxyphenyl, 2-cyano-4-fluorophenyl, 2,4-dimethoxy-phenyl, 2,4- difluorophenyl, 4-isopropoxyphenyl, 2,4-di-trifluoromethylphenyl, A-n-butoxy-2- methylphenyl, 2-methylphenyl, 4-benzyloxy-2-methylphenyl, 4-chloro-2-methoxyphenyl, benzo
  • B, taken together with R 5 , R 6 and R 7 is selected from napthalen-1-yl and naphthalen-2-yl. In some embodiments of the invention, B, taken together with R 5 , R 6 and R 7 , is benzothiophene or benzofuran. In some embodiments, B, taken together with R 5 , R 6 and R 7 , is selected from benzothio-phene-2-yl and benzofuran-2-yl. In some embodiments of the invention, B is indole. In some embodiments, B is N-alkyl substituted indole.
  • B taken together with R 5 , R 6 and R 7 , is selected from l-methylindol-2-yl, l-methylindol-5-yl, 5-methoxyindol-2- yl, and l-methyl-5-methoxyindol-2-yl.
  • B is pyridine.
  • B, taken together with R 5 , R 6 and R 7 is selected from 5- methylpyridine-2-yl, 5-chloropyridin-2-yl and 5-trifluoromethylpyridin-2-yl.
  • B, taken together with R 5 , R 6 and R 7 is selected from quinoline, quinazoline and optionally substituted pyridazine. In some embodiments, B, taken together with R 5 , R 6 and R 7 , is selected from quinolin-2-yl, quinazolin-2-yl, 6- trifluoromethylpyridazin-3-yl. In some embodiments, B, taken together with R 5 , R 6 and R 7 , is l-methylindazol-5-yl. In some embodiments, B, taken together with R 5 , R 6 and R 7 , is 5-methylbenzisoxazol-2-yl.
  • B, taken together with R 5 , R 6 and R 7 is 4-methylcyclohex-l-enyl. In some embodiments, B, taken together with R 5 , R 6 and R 7 , is 4-methylcyclohex- 1 -yl.
  • the compound is selected from one of the following:
  • the pharmaceutically acceptable salt form comprises an HCl salt.
  • the compound is selected from the group comprising:
  • the pharmaceutically acceptable salt form comprises an HCl salt.
  • the compound is selected from the group comprising:
  • the pharmaceutically acceptable salt form comprises an HCl salt.
  • the compound is selected from the group comprising:
  • the pharmaceutically acceptable salt form comprises an HCl salt.
  • a pharmaceutical composition comprising a compound as described herein, and a pharmaceutically acceptable carrier, excipient or diluent therefore.
  • a method of treating obesity comprising administering to a patient in need of obesity reduction an obesity-reducing effective amount of a compound as described herein.
  • a method of treating anxiety comprising administering to a patient in need of such treatment an effective amount of a compound as described herein.
  • a method of treating depression comprising administering to a patient in need of such treatment an effective amount of a compound as described herein.
  • a method of treating a disease or condition which is susceptible to treatment with an MCHi receptor modulator comprising administering to a patient in need thereof a therapeutically effective amount of a compound as described herein.
  • Alkyl is intended to include linear, branched, or cyclic hydrocarbon structures and combinations thereof. When not otherwise restricted, the term refers to alkyl of 20 or fewer carbons. Lower alkyl refers to alkyl groups of 1, 2, 3, 4, 5 and 6 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s-and t-butyl and the like. Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of 3, 4, 5, 6, 7, and 8 carbon atoms.
  • cycloalkyl groups examples include c-propyl, c-butyl, c-pentyl, norbornyl, adamantyl and the like.
  • Other examples of cycloalkyl groups include c-propenyl, c-butenyl, c-pentenyl, and c-hexenyl.
  • Ci to C 20 Hydrocarbon includes alkyl, cycloalkyl, alkenyl, alkynyl, aryl and combinations thereof. Examples include benzyl, phenethyl, cyclohexylmethyl, camphoryl and naphthylethyl.
  • phenylene refers to ortho, meta or para residues of the formulae:
  • Alkoxy or alkoxyl refers to groups of 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to four carbons.
  • alkoxy also includes methylenedioxy and ethylenedioxy in which each oxygen atom is bonded to the atom, chain or ring from which the methylenedioxy or ethylenedioxy group is pendant so as to form a ring.
  • phenyl substituted by alkoxy may be, for example,
  • Oxaalkyl refers to alkyl residues in which one or more carbons (and their associated hydrogens) have been replaced by oxygen. Examples include methoxypropoxy, 3,6,9- trioxadecyl and the like.
  • the term oxaalkyl is intended as it is understood in the art [see Naming and Indexing of Chemical Substances for Chemical Abstracts, published by the American Chemical Society, ⁇
  • thiaalkyl and azaalkyl refer to alkyl residues in which one or more carbons have been replaced by sulfur or nitrogen, respectively. Examples include ethylaminoethyl and methylthiopropyl.
  • Acyl refers to groups of 1, 2, 3, 4, 5, 6, 7 and 8 carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality.
  • One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl.
  • Examples include formyl, acetyl, propionyl, isobutyryl, t-butoxycarbonyl, benzoyl, benzyloxycarbonyl and the like.
  • Lower-acyl refers to groups containing one to four carbons.
  • Aryl and heteroaryl refer to aromatic or heteroaromatic rings, respectively, as substituents.
  • Heteroaryl contains one, two or three heteroatoms selected from O, N, or S. Both refer to monocyclic 5- or 6-membered aromatic or heteroaromatic rings, bicyclic 9- or 10-membered aromatic or heteroaromatic rings and tricyclic 13- or 14-membered aromatic or heteroaromatic rings.
  • Aromatic 6, 7, 8, 9, 10, 11, 12, 13 and 14-membered carbocyclic rings include, e.g., benzene, naphthalene, indane, tetralin, and fluorene and the 5, 6, 7, 8, 9 and 10-membered aromatic heterocyclic rings include, e.g., imidazole, pyridine, indole, thiophene, benzopyranone, thiazole, furan, benzimidazole, quinoline, isoquinoline, quinoxaline, pyrimidine, pyrazine, tetrazole and pyrazole.
  • Arylalkyl means an alkyl residue attached to an aryl ring. Examples are benzyl, phenethyl and the like.
  • Substituted alkyl, aryl, cycloalkyl, heterocyclyl etc. refer to alkyl, aryl, cycloalkyl, or heterocyclyl wherein up to three H atoms in each residue are replaced with alkyl, halogen, haloalkyl, hydroxy, loweralkoxy, carboxy, carboalkoxy (also referred to as alkoxy carbonyl), carboxamido (also referred to as alkylaminocarbonyl), cyano, carbonyl, nitro, amino, alkylamino, dialkylamino, mercapto, alkylthio, sulfoxide, sulfone, acylamino, amidino, phenyl, benzyl, heteroaryl, phenoxy, benzyloxy, or heteroaryloxy.
  • halogen means fluorine, chlorine, bromine or iodine.
  • prodrug refers to a compound that is made more active in vivo. Commonly the conversion of prodrug to drug occurs by enzymatic processes in the liver or blood of the mammal. Many of the compounds of the invention may be chemically modified without absorption into the systemic circulation, and in those cases, activation in vivo may come about by chemical action (as in the acid-catalyzed cleavage in the stomach) or through the intermediacy of enzymes and microflora in the gastrointestinal GI tract.
  • the compounds of this invention can exist in radiolabeled form, i.e., the compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Radioisotopes of hydrogen, carbon, phosphorous, fluorine, iodine and chlorine include 3 H, 14 C, 35 S, 18 F, 32 P, 33 P, 125 I, and 36 Cl, respectively. Compounds that contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of this invention. Radiolabeled compounds described herein and prodrugs thereof can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabeled compounds can be prepared by carrying out the procedures disclosed in the Examples and Schemes by substituting a readily available radiolabeled reagent for a non-radio labeled reagent.
  • the terms "methods of treating or preventing” mean amelioration, prevention or relief from the symptoms and/or effects associated with lipid disorders.
  • preventing refers to administering a medicament beforehand to forestall or obtund an acute episode or, in the case of a chronic condition to diminish the likelihood or seriousness of the condition.
  • prevent is not an absolute term.
  • reference to “treatment” of a patient is intended to include prophylaxis.
  • mammal is used in its dictionary sense.
  • the term “mammal” includes, for example, mice, hamsters, rats, cows, sheep, pigs, goats, and horses, monkeys, dogs (e.g., Canis familiaris), cats, rabbits, guinea pigs, and primates, including humans.
  • Compounds described herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms.
  • Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-.
  • the present invention is meant to include all such possible isomers, as well as mixtures thereof, including racemic and optically pure forms.
  • Optically active (R)- and (S)-, (-)- and (+)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques.
  • the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
  • a compound As used herein, and as would be understood by the person of skill in the art, the recitation of “a compound” is intended to include salts, solvates and inclusion complexes of that compound as well as any stereoisomeric form, or a mixture of any such forms of that compound in any ratio.
  • a compound as described herein, including in the contexts of pharmaceutical compositions, methods of treatment, and compounds per se is provided as the salt form.
  • the salt is a hydrochloride salt.
  • the term "enantiomeric excess" is well known in the art and is defined for a resolution of ab into a + b as
  • enantiomeric excess is related to the older term “optical purity” in that both are measures of the same phenomenon.
  • the value of ee will be a number from 0 to 100, zero being racemic and 100 being pure, single enantiomer.
  • a compound which in the past might have been called 98% optically pure is now more precisely described as 96% ee; in other words, a 90% ee reflects the presence of 95% of one enantiomer and 5% of the other in the material in question.
  • a protecting group refers to a group which is used to mask a functionality during a process step in which it would otherwise react, but in which reaction is undesirable.
  • the protecting group prevents reaction at that step, but may be subsequently removed to expose the original functionality. The removal or "deprotection” occurs after the completion of the reaction or reactions in which the functionality would interfere.
  • Me, Et, Ph, Tf, Ts and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, toluenesulfonyl and methanesulfonyl respectively.
  • a comprehensive list of abbreviations utilized by organic chemists appears in the first issue of each volume of the Journal of Organic Chemistry. The list, which is typically presented in a table entitled “Standard List of Abbreviations" is incorporated herein by reference.
  • a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt or solvate thereof, together with one or more pharmaceutically carriers thereof and optionally one or more other therapeutic ingredients.
  • the carrier(s) must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular), rectal and topical (including dermal, buccal, sublingual and intraocular) administration.
  • the most suitable route may depend upon the condition and disorder of the recipient.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association a compound of formula I or a pharmaceutically acceptable salt or solvate thereof ("active ingredient”) with the carrier, which constitutes one or more accessory ingredients.
  • active ingredient a compound of formula I or a pharmaceutically acceptable salt or solvate thereof
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein.
  • the pharmaceutical compositions may include a "pharmaceutically acceptable inert carrier", and this expression is intended to include one or more inert excipients, which include starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, disintegrating agents, and the like. If desired, tablet dosages of the disclosed compositions may be coated by standard aqueous or nonaqueous techniques, "Pharmaceutically acceptable carrier” also encompasses controlled release means.
  • compositions may also optionally include other therapeutic ingredients, anti-caking agents, preservatives, sweetening agents, colorants, flavors, desiccants, plasticizers, dyes, and the like. Any such optional ingredient must be compatible with the compound of formula I to insure the stability of the formulation.
  • the composition may contain other additives as needed, including for example lactose, glucose, fructose, galactose, trehalose, sucrose, maltose, raffmose, maltitol, melezitose, stachyose, lactitol, palatinite, starch, xylitol, mannitol, myoinositol, and the like, and hydrates thereof, and amino acids, for example alanine, glycine and betaine, and peptides and proteins, for example albumen.
  • additives including for example lactose, glucose, fructose, galactose, trehalose, sucrose, maltose, raffmose, maltitol, melezitose, stachyose, lactitol, palatinite, starch, xylitol, mannitol, myoinositol, and the like, and hydrates thereof, and amino acids,
  • excipients for use as the pharmaceutically acceptable carriers and the pharmaceutically acceptable inert carriers and the aforementioned additional ingredients include, but are not limited to binders, fillers, disintegrants, lubricants, anti-microbial agents, and coating agents.
  • the dose range for adult humans is generally from 0.005 mg to 10 g/day orally. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of compound of formula I which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the precise amount of compound administered to a patient will be the responsibility of the attendant physician. However, the dose employed will depend on a number of factors, including the age and sex of the patient, the precise disorder being treated, and its severity.
  • a dosage unit (e.g. an oral dosage unit) can include from, for example, 1 to 30 mg, 1 to 40 mg, 1 to 100 mg, 1 to 300 mg, 1 to 500 mg, 2 to 500 mg, 3 to 100 mg, 5 to 20 mg, 5 to 100 mg (e.g.
  • the agents can be administered, e.g., by intravenous injection, intramuscular injection, subcutaneous injection, intraperitoneal injection, topical, sublingual, intraarticular (in the joints), intradermal, buccal, ophthalmic (including intraocular), intranasaly (including using a cannula), or by other routes.
  • the agents can be administered orally, e.g., as a tablet or cachet containing a predetermined amount of the active ingredient, gel, pellet, paste, syrup, bolus, electuary, slurry, capsule, powder, granules, as a solution or a suspension in an aqueous liquid or a non-aqueous liquid, as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion, via a micellar formulation (see, e.g. WO 97/11682) via a liposomal formulation (see, e.g., EP 736299,WO 99/59550 and WO 97/13500), via formulations described in WO 03/094886 or in some other form.
  • a micellar formulation see, e.g. WO 97/11682
  • a liposomal formulation see, e.g., EP 736299,WO 99/59550 and WO 97/13500
  • the agents can also be administered transdermally (i.e. via reservoir-type or matrix-type patches, microneedles, thermal poration, hypodermic needles, iontophoresis, electroporation, ultrasound or other forms of sonophoresis, jet injection, or a combination of any of the preceding methods (Prausnitz et al. 2004, Nature Reviews Drug Discovery 3:115)).
  • the agents can be administered locally, for example, at the site of injury to an injured blood vessel.
  • the agents can be coated on a stent.
  • the agents can be administered using high- velocity transdermal particle injection techniques using the hydrogel particle formulation described in U.S. 20020061336. Additional particle formulations are described in WO 00/45792, WO 00/53160, and WO 02/19989.
  • WO 96/11705 provides formulations suitable for transdermal administration.
  • the agents can be administered in the form a suppository or by other vaginal or rectal means.
  • the agents can be administered in a transmembrane formulation as described in WO 90/07923.
  • the agents can be administered non-invasively via the dehydrated particles described in U.S. 6,485,706.
  • the agent can be administered in an enteric-coated drug formulation as described in WO 02/49621.
  • the agents can be administered intranasaly using the formulation described in U.S. 5,179,079.
  • Formulations suitable for parenteral injection are described in WO 00/62759.
  • the agents can be administered using the casein formulation described in U.S. 20030206939 and WO 00/06108.
  • the agents can be administered using the particulate formulations described in U.S. 20020034536.
  • the agents can be administered by pulmonary route utilizing several techniques including but not limited to intratracheal instillation (delivery of solution into the lungs by syringe), intratracheal delivery of liposomes, insufflation (administration of powder formulation by syringe or any other similar device into the lungs) and aerosol inhalation.
  • Aerosols e.g., jet or ultrasonic nebulizers, metered-dose inhalers (MDIs), and dry-Powder inhalers (DPIs)
  • MDIs metered-dose inhalers
  • DPIs dry-Powder inhalers
  • Aerosol formulations are stable dispersions or suspensions of solid material and liquid droplets in a gaseous medium and can be placed into pressurized acceptable propellants, such as hydrofluoroalkanes (HFAs, i.e. HFA- 134a and HFA-227, or a mixture thereof), dichlorodifluoromethane (or other chlorofluorocarbon propellants such as a mixture of Propellants 11, 12, and/or 114), propane, nitrogen, and the like.
  • HFAs hydrofluoroalkanes
  • HFA- 134a and HFA-227 or a mixture thereof
  • dichlorodifluoromethane or other chlorofluorocarbon propellants such as a mixture of Propellants 11, 12, and/or 114
  • propane nitrogen, and the like.
  • Pulmonary formulations may include permeation enhancers such as fatty acids, and saccharides, chelating agents, enzyme inhibitors (e.g., protease inhibitors), adjuvants (e.g., glycocholate, surfactin, span 85, and nafamostat), preservatives (e.g., benzalkonium chloride or chlorobutanol), and ethanol (normally up to 5% but possibly up to 20%, by weight). Ethanol is commonly included in aerosol compositions as it can improve the function of the metering valve and in some cases also improve the stability of the dispersion. Pulmonary formulations may also include surfactants which include but are not limited to bile salts and those described in U.S.
  • the surfactants described in U.S. 6,524,557 e.g., a Cs-Ci 6 fatty acid salt, a bile salt, a phospholipid, or alkyl saccharide are advantageous in that some of them also reportedly enhance absorption of the compound in the formulation.
  • dry powder formulations comprising a therapeutically effective amount of active compound blended with an appropriate carrier and adapted for use in connection with a dry-Powder inhaler.
  • Absorption enhancers which can be added to dry powder formulations of the present invention include those described in U.S. 6,632,456.
  • WO 02/080884 describes new methods for the surface modification of powders. Aerosol formulations may include U.S.
  • Pulmonary formulations containing stable glassy state powder are described in U.S. 20020141945 and U.S. 6,309,671.
  • Other aerosol formulations are described in EP 1338272A1 WO 90/09781, U. S. 5,348,730, U.S. 6,436,367, WO 91/04011, and U.S. 6,294,153 and U.S. 6,290,987 describes a liposomal based formulation that can be administered via aerosol or other means.
  • Powder formulations for inhalation are described in U.S. 20030053960 and WO 01/60341.
  • the agents can be administered intranasally as described in U.S. 20010038824.
  • Solutions of medicament in buffered saline and similar vehicles are commonly employed to generate an aerosol in a nebulizer.
  • Simple nebulizers operate on Bernoulli's principle and employ a stream of air or oxygen to generate the spray particles.
  • More complex nebulizers employ ultrasound to create the spray particles. Both types are well known in the art and are described in standard textbooks of pharmacy such as Sprawls' American Pharmacy and Remington's The Science and Practice of Pharmacy.
  • Other devices for generating aerosols employ compressed gases, usually hydro fluorocarbons and chlorofluorocarbons, which are mixed with the medicament and any necessary excipients in a pressurized container, these devices are likewise described in standard textbooks such as Sprawls and Remington.
  • the agent can be incorporated into a liposome to improve half-life.
  • the agent can also be conjugated to polyethylene glycol (PEG) chains.
  • PEG polyethylene glycol
  • Methods for pegylation and additional formulations containing PEG-conjugates i.e. PEG-based hydrogels, PEG modified liposomes
  • the agent can be administered via a nanocochleate or cochleate delivery vehicle (BioDelivery Sciences International).
  • the agents can be delivered transmucosally (i.e. across a mucosal surface such as the vagina, eye or nose) using formulations such as that described in U.S. 5,204,108.
  • the agents can be formulated in microcapsules as described in WO 88/01165.
  • the agent can be administered intra-orally using the formulations described in U.S. 20020055496, WO 00/47203, and U.S. 6,495,120.
  • the agent can be delivered using nanoemulsion formulations described in WO 01/91728A2.
  • Table 1 lists compounds representative of embodiments of the invention.
  • compounds of formula I may be prepared by the methods illustrated in the general reaction schemes as, for example, described below, or by modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants that are in themselves known, but are not mentioned here.
  • Compounds of formula 2 (wherein Z is chlorine, bromine or iodine) can be prepared by treating compounds of formula 1 with NaNO 2 in acetic acid at room temperature.
  • compounds of formula 2 can be prepared by treatment of amino indazoles 3 with NaNO 2 and copper halide.
  • compounds of formula 2 can be prepared by treatment of aldehydes 4 with hydrazine under heated conditions.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N,N-dimethylformamide (DMF), dimethylsulfoxide (DMSO), acetonitrile and tetrahydrofuran (THF).
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N,N- dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • compounds of formula 8 can be treated under acidic reaction conditions to provide compounds of formula 9.
  • compounds of formula 8 can be treated with an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO to give compounds of formula 9.
  • an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO
  • Treatment of compounds 9 with amines 10 and a reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, or picoline-borane complex can provide compounds of formula 11.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N, ⁇ /-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • compounds of formula 8 can be treated with diamines 14 (wherein R 10 , R 11 , R 12 are each independently H or alkyl) and trimethylaluminum to provide compounds of formula 15.
  • compounds of formula 18 can be be treated with acetic anhydride under heated conditions followed by methanol and water or methanol and sodium hydroxide under ambient to heated conditions to provide compounds of formula 19.
  • compounds of formula 21 can be heated under acid conditions to provide compounds of formula 19.
  • Compounds of formula 6 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8-hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and 4- (benzyloxy)pyridin-2(lH)-one to give compounds of formula 23.
  • a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8-hydroxyquinoline
  • a base such as potassium carbonate, cesium carbonate or potassium phosphate and 4- (benzyloxy)pyridin-2(lH)-one
  • compounds of formula 23 can be treated with hydrogen and a catalyst such as palladium on carbon to provide compounds of formula 24.
  • the hydroxyl group on compounds of formula 24 can be converted to an appropriate activating group to give compounds of formula 25.
  • compounds of formula 24 can be treated with trifluoromethylsulfonic anhydride or JV-phenyl trifluoromethanesulfonamide and a base such as pyridine or lithium bis(trimethylsilyl)amide under cooled conditions to give compounds of formula 25.
  • compounds of formula 25 (wherein Z 6 is triflate) can be be treated with hexaalkylditin and palladium(O) under heated conditions to give compounds of formula 25, wherein Z 6 is trialkylstannane.
  • compounds of formula 8 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and 4-(benzyloxy)pyridin-2(lH)-one to give compounds of formula 27.
  • a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline
  • a base such as potassium carbonate, cesium carbonate or potassium phosphate and 4-(benzyloxy)pyridin-2(lH)-one
  • compounds of formula 27 can be treated with an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO to give compounds of formula 28.
  • an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO
  • compounds of formula 28 Treatment of compounds 28 with amines 10 and a reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, or picoline-borane complex can provide compounds of formula 23, which may be converted to compounds of formula 26 as described above.
  • compounds of formula 32 can be made starting from compounds of formula 29.
  • Compounds of formula 29 (wherein Z 7 is an activating group such as fluorine, chlorine, bromine or iodine) can be treated under heated conditions in a solvent such as DMF with a base such as sodium carbonate or cesium carbonate and 4-(benzyloxy)pyridin- 2(lH)-one to give a compound of formula 30.
  • compound 30 can be treated under reducing conditions such as SnCl 2 , iron powder and NH 4 Cl, or palladium on carbon under a hydrogen atmosphere to provide a compound of formula 31.
  • Treatment of compound 31 with NaNO 2 in acetic acid at room temperature can provide a compound of formula 32.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N, ⁇ /-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • Compounds of formula 23 can be converted to compounds of formula 26 as described above.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to ⁇ /,iV-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • compounds of formula 6 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and compounds of formula 19 to give compounds of formula 26.
  • a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and compounds of formula 19 to give compounds of formula 26.
  • compounds of formula 8 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and compounds of formula 19 to give compounds of formula 34.
  • a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and compounds of formula 19 to give compounds of formula 34.
  • compounds of formula 34 can be treated with an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO to give compounds of formula 35.
  • an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO
  • compounds of formula 35 Treatment of compounds 35 with amines 10 and a reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, or picoline-borane complex can provide compounds of formula 26.
  • compounds of formula 34 can b bee ttrreeaatteedd wwiitthh ddiiaammiinneess 1144 ((wwhheerreeiinn RR 1100 ,, RR 1111 ,, RR 1122 aarree eeaacch independently H or alkyl) and trimethylaluminum to provide compounds of formula 36.
  • compounds of formula 39 can be made starting from compounds of formula 29.
  • Compounds of formula 29 (wherein Z 7 is an activating group such as fluorine, chlorine, bromine or iodine) can be treated under heated conditions in a solvent such as DMF with a base such as sodium carbonate and compounds of formula 19 to give compounds of formula 37.
  • compounds of formula 37 can be treated under reducing conditions such as SnCl 2 , iron powder and NH 4 Cl, or palladium on carbon under a hydrogen atmosphere to provide compounds of formula 38.
  • Treatment of compounds of formula 38 with NaNO 2 in acetic acid at room temperature can provide compounds of formula 39.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N, ⁇ /-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N,N- dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • Compounds of formula 34 can be converted to compounds of formula 26 as described above.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N, ⁇ /-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride.
  • Typical solvents include but are not limited to N, ⁇ /-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran.
  • Treatment of compounds 42 with amines 10 and a Lewis acid such as lithium perchlorate in a sovent such as tetrahyrdrofuran can provide compounds of formula 43.
  • compounds of formula 26 can be treated with hydrogen gas and a catalyst such as palladium on carbon to provide compounds of formula 44, wherein B is fully saturated cycloalkyl.
  • A Water with 0.05% Trifluoroacetic Acid
  • A Water with 0.05% Trifluoroacetic Acid
  • a solution OfNaNO 2 (0.20 g, 2.8 mmol) in H 2 O (5 mL) was cooled in a wet ice bath and treated with a solution of l-(2-(pyrrolidin-l-yl)ethyl)-lH-indazol-5-amine (0.65 g, 2.8 mmol) in 48% aqueous HBr (2 mL).
  • the resulting mixture was added to a pre-heated solution of CuBr (0.49 g, 3.4 mmol) in 48% aqueous HBr (2 mL) at 100 0 C. After stirring at 100 0 C for 15 min, the dark mixture was allowed to cool.
  • the reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml), washed with water (25 mL) and brine (25 mL), dried (Na 2 SO 4 ), and concentrated under reduced pressure.
  • the material was purified by column chromatography (silica gel, 9:1 CH 2 Cl 2 ZMeOH to 90:9:1 CH 2 Cl 2 /Me0H /cone. NH 4 OH).
  • the dried residue 160 mg, 0.35 mmol
  • the reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml), washed with water (25 mL) and brine (25 mL), dried (Na 2 SO 4 ), and concentrated under reduced pressure.
  • the material was purified by column chromatography (silica gel, 9:1 CH 2 Cl 2 /Me0H to 90:9:1 CH 2 Cl 2 /Me0H /cone. NH 4 OH).
  • the dried residue (197 mg, 0.41 mmol) was dissolved in dichloromethane (10 mL) and HCl (1.25 M solution in methanol, 0.36 mL, 0.45 mmol) was added.
  • the reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml), washed with water (25 mL) and brine (25 mL), dried (Na 2 SO 4 ), and concentrated under reduced pressure.
  • the material was purified by column chromatography (silica gel, 9:1 CH 2 Cl 2 /Me0H to 90:9:1 CH 2 Cl 2 /Me0H /cone. NH 4 OH).
  • the dried residue (198 mg, 0.43 mmol) was dissolved in dichloromethane (2 mL), and HCl (1.25 M solution in methanol, 0.38 mL, 0.47 mmol) was added.
  • the reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml) and washed with water (25 mL), brine (25 mL), dried (Na 2 SO 4 ), and concentrated under reduced pressure.
  • the material was purified by column chromatography (silica gel, 9:1 CH 2 Cl 2 MeOH to 90:9:1 CH 2 Cl 2 MeOH /cone. NH 4 OH). The dried residue (233 mg, 0.48 mmol) was dissolved in dichloromethane
  • the mixture was heated to 70 0 C for 20 h. Upon cooling, the mixture was diluted with THF, filtered through celite and rinsed with THF/methylene chloride. The organics were concentrated and purified by flash chromatography (40 g ISCO column eluting with methylene chloride and methanol/ammonia (10:1); 100% methylene chloride to 15% methanol/ammonia over 35 min at 40 mL/min) then further purified by Preparatory HPLC. The appropriate fractions were concentrated and partitioned between methylene chloride and Na 2 COs. The organics were dried with Na 2 SO 4 , filtered, and concentrated to provide the free base.
  • Cell membrane homogenates (5 ⁇ g protein) were incubated for 60 min at 22°C with 0.1 nM [ 125 I][Phe 13 ,Tyr 19 ]-MCH in the absence or presence of the test compound in a buffer containing 25 mM Hepes/Tris (pH 7.4), 5 mM MgCl 2 , 1 niM CaCl 2 and 0.5% bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • the samples were filtered rapidly under vacuum through glass fiber filters (GF/B, Packard) and rinsed several times with an ice-cold buffer containing 25 mM Hepes/Tris (pH 7.4), 500 mM NaCl, 5 mM MgCl 2 , 1 mM CaCl 2 and 0.1% BSA using a 96- sample cell harvester (Unifilter, Packard).
  • the filters were dried, then counted for radioactivity in a scintillation counter (Topcount, Packard) using a scintillation cocktail (Microscint 0, Packard).
  • the results are expressed as a percent inhibition of the control radioligand specific binding.
  • the IC50 value concentration causing a half-maximal inhibition of control specific binding
  • Hill coefficient (TI H ) were determined by non- linear regression analysis of the competition curve using Hill equation curve fitting.
  • the inhibition constant (K 1 ) was calculated from the Cheng Prusoff equation:
  • Nonspecific binding was determined in the presence of 50 ⁇ M l-(5-(4- cyanophenyl)bicyclo[3.1.0]hexan-2-yl)-3-(4-fluoro-3-(trifluoromethyl)phenyl)- 1 -(3-(4- methylpiperazin-l-yl)propyl)urea. Following incubation, the samples were filtered rapidly under vacuum through Skatron 11731 filters, pre-soaked in 0.5% polyethylenimine, and washed with ice-cold 50 niM Tris-HCl buffer, pH 7.4, (wash setting 9,9,0) using a Skatron cell harvester. The filters were counted for radioactivity in a liquid scintillation counter (Tri-Carb 2100TR, Packard) using a scintillation cocktail (Ultima Gold MV, Perkin Elmer).
  • Tri-Carb 2100TR Tri-Carb 2100TR, Packard
  • the results are expressed as a percent inhibition of the control radioligand specific binding.
  • the IC50 value concentration causing a half-maximal inhibition of control specific binding
  • Hill coefficient (TI H ) were determined by non- linear regression analysis of the competition curve using Hill equation curve fitting.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Pain & Pain Management (AREA)
  • Psychiatry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Saccharide Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)

Abstract

5-pyridinone substituted indazoles of the formula and methods of their use are presented.

Description

5-PYRIDINONE SUBSTITUTED INDAZOLES
CROSS-REFERENCE TORELATED APPLICATIONS
[0001] This application claims priority from U.S. Provisional Application Serial Number 60/951,201, filed July 21, 2007, the entire contents of which are incorporated herein by reference.
FIELD OF THE INVENTION
[0002] The invention relates to human melanin- concentrating hormone (MCHi) receptor- selective antagonist 5-pyridinone substituted indazoles that are useful for treating obesity, to pharmaceutical compositions comprising these compounds, and to methods for the treatment of obesity, anxiety, depression, and psychiatric disorders in a mammal.
BACKGROUND OF THE INVENTION
[0003] Obesity and the multitude of co -morbidities associated with obesity such as diabetes, dyslipidemia, coronary heart disease, and certain cancers are a major concern for public health. The currently available pharmaceutical therapies for the treatment of obesity have limited efficacy and side effects that limit their use. Thus, there is a significant medical need for better pharmacotherapy for obesity.
[0004] Melanin-concentrating hormone (MCH) has been identified as an orexigenic peptide that exerts an effect on food intake and body weight regulation. MCH is a cyclic 19 amino acid neuropeptide expressed in the zona incerta and lateral hypothalamus in response to both energy restriction and leptin deficiency. MCH is known to stimulate feeding when injected into the lateral ventricle of rats and the mRNA for MCH is upregulated in the hypothalamus of genetically obese mice (ob/ob) and in fasted control and ob/ob animals. In addition, animals treated with MCH show increases in glucose, insulin and leptin levels, mimicking human metabolic syndrome (Gomori, A. Chronic infusion of MCH causes obesity in mice Am. J. Physiol. Endocrinol. Metab. 284, E583, 2002). Mice lacking MCH are hypophagic and lean with increased metabolic rate, whereas animals over-expressing MCH gain excess weight on both standard and high fat diets. MCH is thought to have effects on other nervous system functions as well (Rocksz, L. L. Biological Examination of Melanin Concentrating Hormone 1 : Multi-tasking from the hypothalamus Drug News Perspect 19(5), 273, 2006). An orphan G-protein coupled receptor (GPCR) was recently identified as a receptor for MCH. Disruption of the binding between MCH and the MCH receptor, i.e. MCH antagonism, may thus be used to counteract the effects of MCH (McBriar, M. D. Recent advances in the discovery of melanin-concentrating hormone receptor antagonists Curr. Opin. Drug Disc. & Dev. 9(4), 496, 2006).
SUMMARY OF THE INVENTION
[0005] There is provided, in accordance with an embodiment of the invention, A compound of formula I:
Figure imgf000003_0001
wherein n is 0 or 1; R is NR1R2, wherein R1 and R2 are each independently H and optionally substituted alkyl, or R1 and R2, together with the N atom to which they are attached, form a 4-7 membered optionally substituted non-aromatic heterocyclic ring which optionally contains 1 or 2 heteroatoms in addition to the N atom shown; R3 and R4 are each independently H or alkyl; or R may combine with either R3 or R4 to form an optionally substituted pyrrolidin-2-yl; B is selected from ary, heteroaryl, and cycloalkyl; R5, R6, R7 are each independently selected from H, -OH, -O-alkyl, alkyl, halo, -CF3, and -CN, -O-aryl, heteroaryl, and hetercyclyl; and R14 is H or -OH; and provided that when n is 0, R3 and R4 are H and R is pyrrolidin-1-yl, then (a) when B is a 5-7-membered monocyclic aromatic heterocycle, at least one heteroatom of said monocyclic aromatic heterocycle is adjacent to the position where B attaches to the pyridinone moiety, and (b) when B is phenyl which is substituted at the 2-position by methoxy or at the 3 -position by methyl, there is at least one additional substituent on the phenyl ring. In some embodiments, when n is 0, R3 and R4 are H and R is pyrrolidin-1-yl, then when B is a bicyclic aromatic heterocycle, either (i) at least one heteroatom of said bicyclic aromatic heterocycle is adjacent to the position where B attaches to the pyridinone moiety, or (ii) the ring of said bicyclic aromatic heterocycle which attaches to the pyridinone moiety does not contain a heteroatom.
[0006] In accordance with some embodiments of the invention, R is selected from the group consisting of pyrrolidin-1-yl, 3-hydroxypyrrolidin-l-yl, morpholin-4-yl, 3- hydroxymethylpyrrolidin-1-yl, dimethylamino, piperazin-1-yl, amino, and 2-oxa-5- azabicyclo[2.2.1]heptane-5-yl. In some embodiments, R is selected from S-3- hydroxypyrrolidin- 1 -yl, R-3 -hydroxypyrrolidin- 1 -yl, S-3 -hydroxymethylpyrro lidin- 1 -yl, R- 3-hydroxymethylpyrrolidin-l-yl, and (liS^iS^-oxa-S-azabicycloP^.lJheptane-S-yl. In some embodiments, R combines with either R3 or R4 to form pyrrolidin-2-yl. In accordance with some embodiments of the invention, R3 and R4 are both H. In some embodiments, n is 0. In other embodiments, n is 1.
[0007] In some embodiments of the invention, B is phenyl. In some embodiments, B, taken together with R5, R6 and R7, is selected from the group consisting of phenyl, 4-chlorophenyl, 4-fluorophenyl, 3-chlorophenyl, 3-phenoxyphenyl, 4-trifluoromethylphenyl, 3- trifluoromethylphenyl, 2,4-dichlorophenyl, 4-methylphenyl, 4-trifluoromethoxyphenyl, A- cyanophenyl, 4-methoxyphenyl, 2-cyano-4-fluorophenyl, 2,4-dimethoxy-phenyl, 2,4- difluorophenyl, 4-isopropoxyphenyl, 2,4-di-trifluoromethylphenyl, A-n-butoxy-2- methylphenyl, 2-methylphenyl, 4-benzyloxy-2-methylphenyl, 4-chloro-2-methoxyphenyl, benzodioxol-5-yl, 4-methoxy-2-methylphenyl, 2-chloro-4-trifluoromethylphenyl, and A- chloro-2-fluorophenyl, 4-trifluoromethyl-2-fluorophenyl, 4-methoxy-2-fluorophenyl, A- methoxy-2-chlorophenyl, 4-ethoxyphenyl, 4-trifluoromethoxy-2-fluorophenyl, and A- trifluoromethoxy-2-methylphenyl, 4-(piperidine-2-yl)phenyl, and 4-(lH-pyrazol-l- yl)phenyl. In other embodiments of the invention, B, taken together with R5, R6 and R7, is selected from napthalen-1-yl and naphthalen-2-yl. In some embodiments of the invention, B, taken together with R5, R6 and R7, is benzothiophene or benzofuran. In some embodiments, B, taken together with R5, R6 and R7, is selected from benzothio-phene-2-yl and benzofuran-2-yl. In some embodiments of the invention, B is indole. In some embodiments, B is N-alkyl substituted indole. In some embodiments, B, taken together with R5, R6 and R7, is selected from l-methylindol-2-yl, l-methylindol-5-yl, 5-methoxyindol-2- yl, and l-methyl-5-methoxyindol-2-yl. In some embodiments of the invention, B is pyridine. In some embodiments, B, taken together with R5, R6 and R7, is selected from 5- methylpyridine-2-yl, 5-chloropyridin-2-yl and 5-trifluoromethylpyridin-2-yl. In some embodiments of the invention, B, taken together with R5, R6 and R7, is selected from quinoline, quinazoline and optionally substituted pyridazine. In some embodiments, B, taken together with R5, R6 and R7, is selected from quinolin-2-yl, quinazolin-2-yl, 6- trifluoromethylpyridazin-3-yl. In some embodiments, B, taken together with R5, R6 and R7, is l-methylindazol-5-yl. In some embodiments, B, taken together with R5, R6 and R7, is 5-methylbenzisoxazol-2-yl. In some embodiments, B, taken together with R5, R6 and R7, is 4-methylcyclohex-l-enyl. In some embodiments, B, taken together with R5, R6 and R7, is 4-methylcyclohex- 1 -yl.
[0008] In some embodiments of the invention, the compound is selected from one of the following:
Figure imgf000005_0001
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
Figure imgf000009_0001
, and , or a pharmaceutically acceptable salt form of the foregoing. In an embodiment of the invention, the pharmaceutically acceptable salt form comprises an HCl salt.
[0009] In some embodiments of the invention, the compound is selected from the group comprising:
Figure imgf000010_0001
Figure imgf000010_0002
Figure imgf000011_0001
Figure imgf000011_0002
Figure imgf000011_0003
Figure imgf000012_0001
Figure imgf000012_0002
and H3C , or a pharmaceutically acceptable salt form of the foregoing. In an embodiment of the invention, the pharmaceutically acceptable salt form comprises an HCl salt.
[0010] In some embodiments of the invention, the compound is selected from the group comprising:
Figure imgf000013_0001
Figure imgf000013_0002
Figure imgf000014_0001
Figure imgf000014_0002
Figure imgf000014_0003
or a pharmaceutically acceptable salt form of the foregoing. In an embodiment of the invention, the pharmaceutically acceptable salt form comprises an HCl salt.
[0011] In some embodiments of the invention, the compound is selected from the group comprising:
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000016_0001
Figure imgf000016_0002
or a pharmaceutically acceptable salt form of the foregoing. In an embodiment of the invention, the pharmaceutically acceptable salt form comprises an HCl salt.
[0012] There is also provided, in accordance with embodiments of the invention, a pharmaceutical composition comprising a compound as described herein, and a pharmaceutically acceptable carrier, excipient or diluent therefore. [0013] There is also provided, in accordance with embodiments of the invention, a method of treating obesity, comprising administering to a patient in need of obesity reduction an obesity-reducing effective amount of a compound as described herein.
[0014] There is also provided, in accordance with embodiments of the invention, a method of treating anxiety, comprising administering to a patient in need of such treatment an effective amount of a compound as described herein.
[0015] There is also provided, in accordance with embodiments of the invention, a method of treating depression, comprising administering to a patient in need of such treatment an effective amount of a compound as described herein.
[0016] There is also provided, in accordance with embodiments of the invention, a method of treating a disease or condition which is susceptible to treatment with an MCHi receptor modulator, comprising administering to a patient in need thereof a therapeutically effective amount of a compound as described herein.
Definitions
[0017] Throughout this specification the terms and substituents retain their definitions.
[0018] Alkyl is intended to include linear, branched, or cyclic hydrocarbon structures and combinations thereof. When not otherwise restricted, the term refers to alkyl of 20 or fewer carbons. Lower alkyl refers to alkyl groups of 1, 2, 3, 4, 5 and 6 carbon atoms. Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, s-and t-butyl and the like. Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of 3, 4, 5, 6, 7, and 8 carbon atoms. Examples of cycloalkyl groups include c-propyl, c-butyl, c-pentyl, norbornyl, adamantyl and the like. Other examples of cycloalkyl groups include c-propenyl, c-butenyl, c-pentenyl, and c-hexenyl.
[0019] Ci to C20 Hydrocarbon (e.g. Ci, C2, C3, C4, C5, C6, C7, C8, C9, Ci0, Cn, Ci2, Ci3, Ci4, C15, Ci6, Ci7, C18, Ci9, C2o) includes alkyl, cycloalkyl, alkenyl, alkynyl, aryl and combinations thereof. Examples include benzyl, phenethyl, cyclohexylmethyl, camphoryl and naphthylethyl. The term "phenylene" refers to ortho, meta or para residues of the formulae:
Figure imgf000018_0001
[0020] Alkoxy or alkoxyl refers to groups of 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to four carbons. For the purposes of the present patent application alkoxy also includes methylenedioxy and ethylenedioxy in which each oxygen atom is bonded to the atom, chain or ring from which the methylenedioxy or ethylenedioxy group is pendant so as to form a ring. Thus, for example, phenyl substituted by alkoxy may be, for example,
Figure imgf000018_0002
[0021] Oxaalkyl refers to alkyl residues in which one or more carbons (and their associated hydrogens) have been replaced by oxygen. Examples include methoxypropoxy, 3,6,9- trioxadecyl and the like. The term oxaalkyl is intended as it is understood in the art [see Naming and Indexing of Chemical Substances for Chemical Abstracts, published by the American Chemical Society, ^|196, but without the restriction of ]|127(a)], i.e. it refers to compounds in which the oxygen is bonded via a single bond to its adjacent atoms (forming ether bonds). Similarly, thiaalkyl and azaalkyl refer to alkyl residues in which one or more carbons have been replaced by sulfur or nitrogen, respectively. Examples include ethylaminoethyl and methylthiopropyl. [0022] Acyl refers to groups of 1, 2, 3, 4, 5, 6, 7 and 8 carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality. One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl. Examples include formyl, acetyl, propionyl, isobutyryl, t-butoxycarbonyl, benzoyl, benzyloxycarbonyl and the like. Lower-acyl refers to groups containing one to four carbons.
[0023] Aryl and heteroaryl refer to aromatic or heteroaromatic rings, respectively, as substituents. Heteroaryl contains one, two or three heteroatoms selected from O, N, or S. Both refer to monocyclic 5- or 6-membered aromatic or heteroaromatic rings, bicyclic 9- or 10-membered aromatic or heteroaromatic rings and tricyclic 13- or 14-membered aromatic or heteroaromatic rings. Aromatic 6, 7, 8, 9, 10, 11, 12, 13 and 14-membered carbocyclic rings include, e.g., benzene, naphthalene, indane, tetralin, and fluorene and the 5, 6, 7, 8, 9 and 10-membered aromatic heterocyclic rings include, e.g., imidazole, pyridine, indole, thiophene, benzopyranone, thiazole, furan, benzimidazole, quinoline, isoquinoline, quinoxaline, pyrimidine, pyrazine, tetrazole and pyrazole.
[0024] Arylalkyl means an alkyl residue attached to an aryl ring. Examples are benzyl, phenethyl and the like.
[0025] Substituted alkyl, aryl, cycloalkyl, heterocyclyl etc. refer to alkyl, aryl, cycloalkyl, or heterocyclyl wherein up to three H atoms in each residue are replaced with alkyl, halogen, haloalkyl, hydroxy, loweralkoxy, carboxy, carboalkoxy (also referred to as alkoxy carbonyl), carboxamido (also referred to as alkylaminocarbonyl), cyano, carbonyl, nitro, amino, alkylamino, dialkylamino, mercapto, alkylthio, sulfoxide, sulfone, acylamino, amidino, phenyl, benzyl, heteroaryl, phenoxy, benzyloxy, or heteroaryloxy.
[0026] The term "halogen" means fluorine, chlorine, bromine or iodine. [0027] The term "prodrug" refers to a compound that is made more active in vivo. Commonly the conversion of prodrug to drug occurs by enzymatic processes in the liver or blood of the mammal. Many of the compounds of the invention may be chemically modified without absorption into the systemic circulation, and in those cases, activation in vivo may come about by chemical action (as in the acid-catalyzed cleavage in the stomach) or through the intermediacy of enzymes and microflora in the gastrointestinal GI tract.
[0028] In the characterization of some of the substituents, it is recited that certain substituents may combine to form rings. Unless stated otherwise, it is intended that such rings may exhibit various degrees of unsaturation (from fully saturated to fully unsaturated), may include heteroatoms and may be substituted with lower alkyl or alkoxy.
[0029] It will be recognized that the compounds of this invention can exist in radiolabeled form, i.e., the compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Radioisotopes of hydrogen, carbon, phosphorous, fluorine, iodine and chlorine include 3H, 14C, 35S, 18F, 32P, 33P, 125I, and 36Cl, respectively. Compounds that contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of this invention. Radiolabeled compounds described herein and prodrugs thereof can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabeled compounds can be prepared by carrying out the procedures disclosed in the Examples and Schemes by substituting a readily available radiolabeled reagent for a non-radio labeled reagent.
[0030] The terms "methods of treating or preventing" mean amelioration, prevention or relief from the symptoms and/or effects associated with lipid disorders. The term "preventing" as used herein refers to administering a medicament beforehand to forestall or obtund an acute episode or, in the case of a chronic condition to diminish the likelihood or seriousness of the condition. The person of ordinary skill in the medical art (to which the present method claims are directed) recognizes that the term "prevent" is not an absolute term. In the medical art it is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or seriousness of a condition, and this is the sense intended in applicants' claims. As used herein, reference to "treatment" of a patient is intended to include prophylaxis.
[0031] Throughout this application, various references are referred to. Each of the patents, patent applications, patent publications, and references mentioned herein is hereby incorporated by reference in its entirety.
[0032] The term "mammal" is used in its dictionary sense. The term "mammal" includes, for example, mice, hamsters, rats, cows, sheep, pigs, goats, and horses, monkeys, dogs (e.g., Canis familiaris), cats, rabbits, guinea pigs, and primates, including humans.
[0033] Compounds described herein may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms. Each chiral center may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. The present invention is meant to include all such possible isomers, as well as mixtures thereof, including racemic and optically pure forms. Optically active (R)- and (S)-, (-)- and (+)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefmic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers. Likewise, all tautomeric forms are also intended to be included.
[0034] As used herein, and as would be understood by the person of skill in the art, the recitation of "a compound" is intended to include salts, solvates and inclusion complexes of that compound as well as any stereoisomeric form, or a mixture of any such forms of that compound in any ratio. Thus, in accordance with some embodiments of the invention, a compound as described herein, including in the contexts of pharmaceutical compositions, methods of treatment, and compounds per se, is provided as the salt form. In accordance with some embodiments of the invention, the salt is a hydrochloride salt. [0035] The term "enantiomeric excess" is well known in the art and is defined for a resolution of ab into a + b as
Figure imgf000022_0001
[0036] The term "enantiomeric excess" is related to the older term "optical purity" in that both are measures of the same phenomenon. The value of ee will be a number from 0 to 100, zero being racemic and 100 being pure, single enantiomer. A compound which in the past might have been called 98% optically pure is now more precisely described as 96% ee; in other words, a 90% ee reflects the presence of 95% of one enantiomer and 5% of the other in the material in question.
[0037] The configuration of any carbon-carbon double bond appearing herein is selected for convenience only and is not intended to designate a particular configuration; thus a carbon- carbon double bond depicted arbitrarily herein as E may be Z, E, or a mixture of the two in any proportion.
[0038] Terminology related to "protecting", "deprotecting" and "protected" functionalities occurs throughout this application. Such terminology is well understood by persons of skill in the art and is used in the context of processes which involve sequential treatment with a series of reagents. In that context, a protecting group refers to a group which is used to mask a functionality during a process step in which it would otherwise react, but in which reaction is undesirable. The protecting group prevents reaction at that step, but may be subsequently removed to expose the original functionality. The removal or "deprotection" occurs after the completion of the reaction or reactions in which the functionality would interfere. Thus, when a sequence of reagents is specified, as it is in the processes of the invention, the person of ordinary skill can readily envision those groups that would be suitable as "protecting groups". Suitable groups for that purpose are discussed in standard textbooks in the field of chemistry, such as Protective Groups in Organic Synthesis by T.W.Greene [John Wiley & Sons, New York, 1991], which is incorporated herein by reference. Particular attention is drawn to the chapters entitled "Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols" (pages 10-86).
[0039] The abbreviations Me, Et, Ph, Tf, Ts and Ms represent methyl, ethyl, phenyl, trifluoromethanesulfonyl, toluenesulfonyl and methanesulfonyl respectively. A comprehensive list of abbreviations utilized by organic chemists (i.e. persons of ordinary skill in the art) appears in the first issue of each volume of the Journal of Organic Chemistry. The list, which is typically presented in a table entitled "Standard List of Abbreviations" is incorporated herein by reference.
[0040] While it may be possible for compounds of formula I to be administered as the raw chemical, it will often be preferable to present them as part of a pharmaceutical composition. In accordance with an embodiment of the present invention there is provided a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt or solvate thereof, together with one or more pharmaceutically carriers thereof and optionally one or more other therapeutic ingredients. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Furthermore, when reference is made in an independent claim to a compound or a pharmaceutically acceptable salt thereof, it will be understood that claims which depend from that independent claim which refer to such a compound also include pharmaceutically acceptable salts of the compound, even if explicit reference is not made to the salts in the dependent claim.
[0041] The formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular), rectal and topical (including dermal, buccal, sublingual and intraocular) administration. The most suitable route may depend upon the condition and disorder of the recipient. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association a compound of formula I or a pharmaceutically acceptable salt or solvate thereof ("active ingredient") with the carrier, which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
[0042] Formulations suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
[0043] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein. The pharmaceutical compositions may include a "pharmaceutically acceptable inert carrier", and this expression is intended to include one or more inert excipients, which include starches, polyols, granulating agents, microcrystalline cellulose, diluents, lubricants, binders, disintegrating agents, and the like. If desired, tablet dosages of the disclosed compositions may be coated by standard aqueous or nonaqueous techniques, "Pharmaceutically acceptable carrier" also encompasses controlled release means.
[0044] Pharmaceutical compositions may also optionally include other therapeutic ingredients, anti-caking agents, preservatives, sweetening agents, colorants, flavors, desiccants, plasticizers, dyes, and the like. Any such optional ingredient must be compatible with the compound of formula I to insure the stability of the formulation. The composition may contain other additives as needed, including for example lactose, glucose, fructose, galactose, trehalose, sucrose, maltose, raffmose, maltitol, melezitose, stachyose, lactitol, palatinite, starch, xylitol, mannitol, myoinositol, and the like, and hydrates thereof, and amino acids, for example alanine, glycine and betaine, and peptides and proteins, for example albumen.
[0045] Examples of excipients for use as the pharmaceutically acceptable carriers and the pharmaceutically acceptable inert carriers and the aforementioned additional ingredients include, but are not limited to binders, fillers, disintegrants, lubricants, anti-microbial agents, and coating agents.
[0046] The dose range for adult humans is generally from 0.005 mg to 10 g/day orally. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of compound of formula I which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg. The precise amount of compound administered to a patient will be the responsibility of the attendant physician. However, the dose employed will depend on a number of factors, including the age and sex of the patient, the precise disorder being treated, and its severity.
[0047] A dosage unit (e.g. an oral dosage unit) can include from, for example, 1 to 30 mg, 1 to 40 mg, 1 to 100 mg, 1 to 300 mg, 1 to 500 mg, 2 to 500 mg, 3 to 100 mg, 5 to 20 mg, 5 to 100 mg (e.g. 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 450 mg, 500 mg) of a compound described herein.
[0048] For additional information about pharmaceutical compositions and their formulation, see, for example, Remington: The Science and Practice of Pharmacy, 20th Edition, 2000.
[0049] The agents can be administered, e.g., by intravenous injection, intramuscular injection, subcutaneous injection, intraperitoneal injection, topical, sublingual, intraarticular (in the joints), intradermal, buccal, ophthalmic (including intraocular), intranasaly (including using a cannula), or by other routes. The agents can be administered orally, e.g., as a tablet or cachet containing a predetermined amount of the active ingredient, gel, pellet, paste, syrup, bolus, electuary, slurry, capsule, powder, granules, as a solution or a suspension in an aqueous liquid or a non-aqueous liquid, as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion, via a micellar formulation (see, e.g. WO 97/11682) via a liposomal formulation (see, e.g., EP 736299,WO 99/59550 and WO 97/13500), via formulations described in WO 03/094886 or in some other form. The agents can also be administered transdermally (i.e. via reservoir-type or matrix-type patches, microneedles, thermal poration, hypodermic needles, iontophoresis, electroporation, ultrasound or other forms of sonophoresis, jet injection, or a combination of any of the preceding methods (Prausnitz et al. 2004, Nature Reviews Drug Discovery 3:115)). The agents can be administered locally, for example, at the site of injury to an injured blood vessel. The agents can be coated on a stent. The agents can be administered using high- velocity transdermal particle injection techniques using the hydrogel particle formulation described in U.S. 20020061336. Additional particle formulations are described in WO 00/45792, WO 00/53160, and WO 02/19989. An example of a transdermal formulation containing plaster and the absorption promoter dimethylisosorbide can be found in WO 89/04179. WO 96/11705 provides formulations suitable for transdermal administration. The agents can be administered in the form a suppository or by other vaginal or rectal means. The agents can be administered in a transmembrane formulation as described in WO 90/07923. The agents can be administered non-invasively via the dehydrated particles described in U.S. 6,485,706. The agent can be administered in an enteric-coated drug formulation as described in WO 02/49621. The agents can be administered intranasaly using the formulation described in U.S. 5,179,079. Formulations suitable for parenteral injection are described in WO 00/62759. The agents can be administered using the casein formulation described in U.S. 20030206939 and WO 00/06108. The agents can be administered using the particulate formulations described in U.S. 20020034536.
[0050] The agents, alone or in combination with other suitable components, can be administered by pulmonary route utilizing several techniques including but not limited to intratracheal instillation (delivery of solution into the lungs by syringe), intratracheal delivery of liposomes, insufflation (administration of powder formulation by syringe or any other similar device into the lungs) and aerosol inhalation. Aerosols (e.g., jet or ultrasonic nebulizers, metered-dose inhalers (MDIs), and dry-Powder inhalers (DPIs)) can also be used in intranasal applications. Aerosol formulations are stable dispersions or suspensions of solid material and liquid droplets in a gaseous medium and can be placed into pressurized acceptable propellants, such as hydrofluoroalkanes (HFAs, i.e. HFA- 134a and HFA-227, or a mixture thereof), dichlorodifluoromethane (or other chlorofluorocarbon propellants such as a mixture of Propellants 11, 12, and/or 114), propane, nitrogen, and the like. Pulmonary formulations may include permeation enhancers such as fatty acids, and saccharides, chelating agents, enzyme inhibitors (e.g., protease inhibitors), adjuvants (e.g., glycocholate, surfactin, span 85, and nafamostat), preservatives (e.g., benzalkonium chloride or chlorobutanol), and ethanol (normally up to 5% but possibly up to 20%, by weight). Ethanol is commonly included in aerosol compositions as it can improve the function of the metering valve and in some cases also improve the stability of the dispersion. Pulmonary formulations may also include surfactants which include but are not limited to bile salts and those described in U.S. 6,524,557 and references therein. The surfactants described in U.S. 6,524,557, e.g., a Cs-Ci6 fatty acid salt, a bile salt, a phospholipid, or alkyl saccharide are advantageous in that some of them also reportedly enhance absorption of the compound in the formulation. Also suitable in the invention are dry powder formulations comprising a therapeutically effective amount of active compound blended with an appropriate carrier and adapted for use in connection with a dry-Powder inhaler. Absorption enhancers which can be added to dry powder formulations of the present invention include those described in U.S. 6,632,456. WO 02/080884 describes new methods for the surface modification of powders. Aerosol formulations may include U.S. 5,230,884, U.S. 5,292,499, WO 017/8694, WO 01/78696, U.S. 2003019437, U. S. 20030165436, and WO 96/40089 (which includes vegetable oil). Sustained release formulations suitable for inhalation are described in U.S. 20010036481A1, 20030232019A1, and U.S. 20040018243A1 as well as in WO 01/13891, WO 02/067902, WO 03/072080, and WO 03/079885. Pulmonary formulations containing microparticles are described in WO 03/015750, U.S. 20030008013, and WO 00/00176. Pulmonary formulations containing stable glassy state powder are described in U.S. 20020141945 and U.S. 6,309,671. Other aerosol formulations are described in EP 1338272A1 WO 90/09781, U. S. 5,348,730, U.S. 6,436,367, WO 91/04011, and U.S. 6,294,153 and U.S. 6,290,987 describes a liposomal based formulation that can be administered via aerosol or other means. Powder formulations for inhalation are described in U.S. 20030053960 and WO 01/60341. The agents can be administered intranasally as described in U.S. 20010038824.
[0051] Solutions of medicament in buffered saline and similar vehicles are commonly employed to generate an aerosol in a nebulizer. Simple nebulizers operate on Bernoulli's principle and employ a stream of air or oxygen to generate the spray particles. More complex nebulizers employ ultrasound to create the spray particles. Both types are well known in the art and are described in standard textbooks of pharmacy such as Sprawls' American Pharmacy and Remington's The Science and Practice of Pharmacy. Other devices for generating aerosols employ compressed gases, usually hydro fluorocarbons and chlorofluorocarbons, which are mixed with the medicament and any necessary excipients in a pressurized container, these devices are likewise described in standard textbooks such as Sprawls and Remington.
[0052] The agent can be incorporated into a liposome to improve half-life. The agent can also be conjugated to polyethylene glycol (PEG) chains. Methods for pegylation and additional formulations containing PEG-conjugates (i.e. PEG-based hydrogels, PEG modified liposomes) can be found in Harris and Chess, Nature Reviews Drug Discovery 2:214-221 and the references therein. The agent can be administered via a nanocochleate or cochleate delivery vehicle (BioDelivery Sciences International). The agents can be delivered transmucosally (i.e. across a mucosal surface such as the vagina, eye or nose) using formulations such as that described in U.S. 5,204,108. The agents can be formulated in microcapsules as described in WO 88/01165. The agent can be administered intra-orally using the formulations described in U.S. 20020055496, WO 00/47203, and U.S. 6,495,120. The agent can be delivered using nanoemulsion formulations described in WO 01/91728A2.
[0053] Table 1 lists compounds representative of embodiments of the invention. [0054] In general, compounds of formula I may be prepared by the methods illustrated in the general reaction schemes as, for example, described below, or by modifications thereof, using readily available starting materials, reagents and conventional synthesis procedures. In these reactions, it is also possible to make use of variants that are in themselves known, but are not mentioned here.
[0055] Processes for obtaining compounds of formula I are presented below. Other compounds of formula I may be prepared in analogous fashion to those whose synthesis is exemplified herein. The procedures below illustrate such methods. Furthermore, although the syntheses depicted herein may result in the preparation of enantiomers having a particular stereochemistry, included within the scope of the present invention are compounds of formula I in any stereoisomeric form, and preparation of compounds of formula I in stereoisomeric forms other than those depicted herein would be obvious to one of ordinary skill in the chemical arts based on the procedures presented herein.
Synthetic Methods
Scheme 1
Figure imgf000029_0001
1 2
[0056] Compounds of formula 2 (wherein Z is chlorine, bromine or iodine) can be prepared by treating compounds of formula 1 with NaNO2 in acetic acid at room temperature.
Scheme 2
Figure imgf000029_0002
3 2
[0057] Alternatively, compounds of formula 2 can be prepared by treatment of amino indazoles 3 with NaNO2 and copper halide.
Scheme 3
Figure imgf000030_0001
4 2
[0058] Alternatively, compounds of formula 2 can be prepared by treatment of aldehydes 4 with hydrazine under heated conditions.
Scheme 4
Figure imgf000030_0002
[0059] Compounds of formula 2 can be treated with base and compounds of formula 5 (wherein Z2 = halogen, methanesulfonate, toluenesulfonate or the like; n = 2 or 3) under ambient temperature or heated conditions to give compounds of formula 6. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,N-dimethylformamide (DMF), dimethylsulfoxide (DMSO), acetonitrile and tetrahydrofuran (THF).
Scheme 5
Figure imgf000030_0003
R1
Figure imgf000030_0004
[0060] Alternatively, compounds of formula 2 can be treated with base and compounds of formula 7 (wherein Z2 = halogen, methanesulfonate, toluenesulfonate or the like; Y = O, OR9 or H; R8 = an alkyl group, H or a protecting group such as te/t-butyldimethylsilyl; R9 = alkyl; n = 1 or 2) under ambient temperature or heated conditions to give compounds of formula 8. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,N- dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. In the case where Y = OR9, compounds of formula 8 can be treated under acidic reaction conditions to provide compounds of formula 9. In the case where Y = H and R8 = a protecting group, compounds of formula 8 can be treated under appropriate deprotecting conditions to provide compounds of formula 8 wherein R8 = H. In the case where Y = H and R8 = H, compounds of formula 8 can be treated with an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO to give compounds of formula 9. Treatment of compounds 9 with amines 10 and a reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, or picoline-borane complex can provide compounds of formula 11.
Figure imgf000031_0001
[0061] Alternatively, compound 2 can be treated with base and compounds of formula 12 (wherein n = 1 or 2 and Z3 and Z4 = halogen, methanesulfonate, toluenesulfonate or the like) under ambient temperature or heated conditions to give compounds of formula 13. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,Λ/-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Treatment of compounds of formula 13 with amines 10 under ambient temperature or heated conditions can provide compounds of formula 11 wherein n = 2 or 3. Scheme 7
Figure imgf000032_0001
15
[0062] Additionally, in the case where Y = O and R = alkyl, compounds of formula 8 can be treated with diamines 14 (wherein R10, R11, R12 are each independently H or alkyl) and trimethylaluminum to provide compounds of formula 15.
Scheme 8
Figure imgf000032_0002
[0063] Compounds of formula 19 can be prepared by treating compounds of formula 16 (wherein X1 is chlorine, bromine or iodine) with compounds of formula 17 (wherein Z5 = B(OH)2, B(OR13)2, SnR13S or the like and R13 = alkyl), a catalyst such as palladium(O), and a base such as potassium carbonate to give compounds of formula 18. In turn, compounds of formula 18 can be be treated with acetic anhydride under heated conditions followed by methanol and water or methanol and sodium hydroxide under ambient to heated conditions to provide compounds of formula 19.
Scheme 9
Figure imgf000032_0003
Alternatively, compounds of formula 19 can be prepared by treating compounds of formula 20 (wherein X2 is chlorine, bromine or iodine) with compounds of formula 17 (wherein Z5 is B(OH)2, B(OR13)2, SnR13 3 or the like and R13 = alkyl), a catalyst such as palladium(O), and a base such as potassium carbonate to give compounds of formula 21. In turn, compounds of formula 21 can be heated under acid conditions to provide compounds of formula 19.
Figure imgf000033_0001
[0064] Compounds of formula 6 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8-hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and 4- (benzyloxy)pyridin-2(lH)-one to give compounds of formula 23. In turn, compounds of formula 23 can be treated with hydrogen and a catalyst such as palladium on carbon to provide compounds of formula 24. The hydroxyl group on compounds of formula 24 can be converted to an appropriate activating group to give compounds of formula 25. In the case where Z6 is triflate, compounds of formula 24 can be treated with trifluoromethylsulfonic anhydride or JV-phenyl trifluoromethanesulfonamide and a base such as pyridine or lithium bis(trimethylsilyl)amide under cooled conditions to give compounds of formula 25. In the case where Z6 is trialkylstannane, compounds of formula 25 (wherein Z6 is triflate) can be be treated with hexaalkylditin and palladium(O) under heated conditions to give compounds of formula 25, wherein Z6 is trialkylstannane. Treatment of compounds of formula 25 with compounds of formula 17 (wherein Z5 = an appropriately matched activating group, such as B(OH)2, B(OR13)2, SnR13S, halogen or the like and R13 = alkyl), a catalyst such as palladium(O), and a base such as potassium carbonate under heated conditions can provide compounds of formula 26.
[0065] Additionally, compounds of formula 8 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and 4-(benzyloxy)pyridin-2(lH)-one to give compounds of formula 27. In the case where Y = OR9, compounds of formula 27 can be treated under acidic reaction conditions to provide compounds of formula 28. In the case where Y = H and R8 = a protecting group, compounds of formula 27 can be treated under appropriate deprotecting conditions to provide compounds of formula 27 wherein R8 = H. In the case where Y = H and R8 = H, compounds of formula 27 can be treated with an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO to give compounds of formula 28. Treatment of compounds 28 with amines 10 and a reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, or picoline-borane complex can provide compounds of formula 23, which may be converted to compounds of formula 26 as described above.
Figure imgf000035_0001
[0066] Alternatively, compounds of formula 32 can be made starting from compounds of formula 29. Compounds of formula 29 (wherein Z7 is an activating group such as fluorine, chlorine, bromine or iodine) can be treated under heated conditions in a solvent such as DMF with a base such as sodium carbonate or cesium carbonate and 4-(benzyloxy)pyridin- 2(lH)-one to give a compound of formula 30. In turn, compound 30 can be treated under reducing conditions such as SnCl2, iron powder and NH4Cl, or palladium on carbon under a hydrogen atmosphere to provide a compound of formula 31. Treatment of compound 31 with NaNO2 in acetic acid at room temperature can provide a compound of formula 32. Compound 32 can be treated with a base and compounds of formula 5 (wherein Z2 = halogen, methanesulfonate, toluenesulfonate or the like; n = 2 or 3) under ambient temperature or heated conditions to give compounds of formula 23. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,Λ/-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Compounds of formula 23 can be converted to compounds of formula 26 as described above.
Scheme 13
Figure imgf000036_0001
[0067] Alternatively, compound 32 can be treated with base and compounds of formula 7 (wherein Z2 = halogen, methanesulfonate, toluenesulfonate or the like; Y = O, OR9 or H; R8
= an alkyl group, H or a protecting group such as te/t-butyldimethylsilyl; R > 9 . = alkyl; n = 1 or 2) under ambient temperature or heated conditions to give compounds of formula 27. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to ΛζiV-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Compounds of formula 27 can be converted to compounds of formula 26 as described above. Scheme 14
Figure imgf000037_0001
[0068] Alternatively, compound 32 can be treated with base and compounds of formula 12 (wherein n = 1 or 2 and Z3 and Z4 = halogen, methanesulfonate, toluenesulfonate or the like) under ambient temperature or heated conditions to give compounds of formula 33. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to Λ/,iV-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Treatment of compounds of formula 33 with amines 10 under ambient temperature or heated conditions can provide compounds of formula 23 wherein n = 2 or 3, which may be converted to compounds of formula 26 as described above.
Figure imgf000038_0001
[0069] Alternatively, compounds of formula 6 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and compounds of formula 19 to give compounds of formula 26.
Figure imgf000038_0002
[0070] Additionally, compounds of formula 8 can be treated under heated conditions with a catalyst such as copper iodide, a ligand such as trans- 1,2-diaminocyclohexane or 8- hydroxyquinoline, a base such as potassium carbonate, cesium carbonate or potassium phosphate and compounds of formula 19 to give compounds of formula 34. In the case where Y = OR9, compounds of formula 34 can be treated under acidic reaction conditions to provide compounds of formula 35. In the case where Y = H and R8 = a protecting group, compounds of formula 34 can be treated under appropriate deprotecting conditions to provide compounds of formula 34 wherein R8 = H. In the case where Y = H and R8 = H, compounds of formula 34 can be treated with an oxidizing agent such as the Dess-Martin periodane or oxalyl chloride and DMSO to give compounds of formula 35. Treatment of compounds 35 with amines 10 and a reducing agent such as sodium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, or picoline-borane complex can provide compounds of formula 26.
Scheme 17
Figure imgf000039_0001
[0071] Additionally, in the case where Y = O and R8 = alkyl, compounds of formula 34 can b bee ttrreeaatteedd wwiitthh ddiiaammiinneess 1144 ((wwhheerreeiinn RR1100,, RR1111,, RR1122 aarree eeaacch independently H or alkyl) and trimethylaluminum to provide compounds of formula 36.
Scheme 18
Figure imgf000040_0001
[0072] Alternatively, compounds of formula 39 can be made starting from compounds of formula 29. Compounds of formula 29 (wherein Z7 is an activating group such as fluorine, chlorine, bromine or iodine) can be treated under heated conditions in a solvent such as DMF with a base such as sodium carbonate and compounds of formula 19 to give compounds of formula 37. In turn, compounds of formula 37 can be treated under reducing conditions such as SnCl2, iron powder and NH4Cl, or palladium on carbon under a hydrogen atmosphere to provide compounds of formula 38. Treatment of compounds of formula 38 with NaNO2 in acetic acid at room temperature can provide compounds of formula 39. Compounds of formula 39 can be treated with base and compounds of formula 5 (wherein Z2 = halogen, methanesulfonate, toluenesulfonate or the like; n = 2 or 3) under ambient temperature or heated conditions to give compounds of formula 26. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,Λ/-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Scheme 19
Figure imgf000041_0001
[0073] Alternatively, compounds of formula 39 can be treated with base and compounds of formula 7 (wherein Z2 = halogen, methanesulfonate, toluenesulfonate or the like; Y = O, OR9 or H; R8 = an alkyl group, H or a protecting group such as tert-butyldimethylsilyl; R9 = alkyl; n = 1 or 2) under ambient temperature or heated conditions to give compounds of formula 34. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,N- dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Compounds of formula 34 can be converted to compounds of formula 26 as described above.
Figure imgf000041_0002
[0074] Alternatively, compound 39 can be treated with base and compounds of formula 12 (wherein n = 1 or 2 and Z3 and Z4 = halogen, methanesulfonate, toluenesulfonate or the like) under ambient temperature or heated conditions to give compounds of formula 40. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,Λ/-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Treatment of compounds of formula 40 with amines 10 under ambient temperature or heated conditions can provide compounds of formula 26 wherein n = 2 or 3.
Scheme 21
Figure imgf000042_0001
[0075] Compounds of formula 39 (wherein B is aryl or heteroaryl and R5, R6, R7 are each independently selected from H, -OH, -O-alkyl, alkyl, halo, -CF3, and -CN) can be treated with base and compounds of formula 41 (wherein Z8 = halogen, methanesulfonate, toluenesulfonate, 3-nitrobenzenesulfonate or the like) at ambient temperature or under heated conditions to give compounds of formula 42. Typical bases include but are not limited to cesium carbonate, potassium carbonate and sodium hydride. Typical solvents include but are not limited to N,Λ/-dimethylformamide, dimethylsulfoxide, acetonitrile and tetrahydrofuran. Treatment of compounds 42 with amines 10 and a Lewis acid such as lithium perchlorate in a sovent such as tetrahyrdrofuran can provide compounds of formula 43. Scheme 22
Figure imgf000043_0001
[0076] In the case where B contains a single site of unsaturation, compounds of formula 26 can be treated with hydrogen gas and a catalyst such as palladium on carbon to provide compounds of formula 44, wherein B is fully saturated cycloalkyl.
Examples
[0077] Unless otherwise noted, reagents and solvents were used as received from commercial suppliers. Proton nuclear magnetic resonance (NMR) spectra were obtained on Bruker spectrometers at 300, 400 or 500 MHz. Spectra are given in ppm (δ) and coupling constants, J, are reported in Hertz. Tetramethylsilane (TMS) was used as an internal standard. Mass spectra were collected using either a Finnigan LCQ Duo LCMS ion trap electrospray ionization (ESI) or a mass Varian 1200L single quadrapole mass spectrometer (ESI). High performace liquid chromatograph (HPLC) analyses were obtained using a Luna C 18(2) column (5u, 250 x 4.6 mm, Phenomenex) or a Gemini Cl 8 column (5u, 250 x 4.5 mm, Phenomenex) with UV detection at 254 nm using a standard solvent gradient program (Method A, Method B, Method C, Method D, or Method E).
Method A:
Figure imgf000043_0002
A = Water with 0.025% Trifluoroacetic Acid
B = Acetonitrile with 0.025% Trifluoroacetic Acid Method B:
Figure imgf000044_0001
A = Water with 0.05% Trifluoroacetic Acid
B = Acetonitrile with 0.05% Trifluoroacetic Acid
Method C:
Figure imgf000044_0002
A = Water with 0.025% Trifluoroacetic Acid
B = Acetonitrile with 0.025% Trifluoroacetic Acid
Method D:
Figure imgf000044_0003
A = Water with 0.05% Trifluoroacetic Acid
B = Acetonitrile with 0.05% Trifluoroacetic Acid
Method E:
Figure imgf000044_0004
A = Water with 0.025% Trifluoroacetic Acid
B = Acetonitrile with 0.025% Trifluoroacetic Acid
Example 1 Preparation of 4-(Phenyl)- 1 -( 1 -(2-(pyrrolidin- 1 -vDethyl)- lH-indazol-5-yl)pyridin-2( lHVone hydrochloride a) 1 -(2-(Pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-amine Beilstein Registry Number 10008406
Chemical Formula: C]3Hi8N4
Exact Mass: 230.15 Molecular Weight: 230.31
Figure imgf000045_0001
[0078] This compound was prepared in accordance with the procedure of Souers et al, Bioorg. Med. Chem. Lett. 2005, 15, 2752-2757.
b) 5-Bromo- 1 -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazole
Chemical Formula: Ci3H16BrN3
Exact Mass: 293.05 Molecular Weight: 294.19
Figure imgf000045_0002
[0079] A solution OfNaNO2 (0.20 g, 2.8 mmol) in H2O (5 mL) was cooled in a wet ice bath and treated with a solution of l-(2-(pyrrolidin-l-yl)ethyl)-lH-indazol-5-amine (0.65 g, 2.8 mmol) in 48% aqueous HBr (2 mL). The resulting mixture was added to a pre-heated solution of CuBr (0.49 g, 3.4 mmol) in 48% aqueous HBr (2 mL) at 1000C. After stirring at 1000C for 15 min, the dark mixture was allowed to cool. The solids were isolated by filtration, washed with IN NaOH, and dried under vacuum. Purification by flash column chromatography (silica gel, CH2CVMeOH, 95:5 to 90:10 gave the title compound (0.21 g, 25%) as a brown solid: ESI MS m/z 294 [M + H]+.
c) 4-(Benzyloxy)- 1 -( 1 -(2-(pyrrolidin- 1 -yl)ethyl)- l/f-indazol-5-yl)pyridin-2( lH)-one Chemical Formula: C25H26N4O2
Exact Mass: 414.21 Molecular Weight: 414.5
Figure imgf000046_0001
[0080] A suspension of 5-bromo-l-(2-(pyrrolidin-l-yl)ethyl)-l/f-indazole (0.21 g, 0.70 mmol) in 1,4-dioxane (10 niL) stirred under nitrogen was treated sequentially with 4- (benzyloxy)pyridin-2(lH)-one (0.14 g, 0.70 mmol), trans- 1,2-diaminocyclohexane (0.03 mL, 0.2 mmol), CuI (28 mg, 0.15 mmol) and K2CO3 (0.19 g, 1.4 mmol). After stirring overnight at 1100C, the mixture was allowed to cool to room temperature, diluted with CH2Cl2, washed with brine, dried over Na2SO4, filtered and concentrated to dryness. Purification by flash column chromatography (silica gel, CH2CVMeOH, 95:5 to 90:10 gave the title compound (21 mg, 7%) as an off-white powder: 1H NMR (500 MHz, DMSO-J6) δ 8.03 (s, IH), 7.66 (d, J= 1.4 Hz, IH), 7.53 (d, J= 8.8 Hz, IH), 7.43-7.36 (m, 6H), 7.28 (d, J = 7.5 Hz, 1 H), 6.09-6.06 (m, 2H), 5.06 (s, 2H), 4.58-4.55 (m, 2H), 3.03 (br s, 2H), 2.61 (br s, 4H), 1.81 (br s, 4H); ESI MS m/z 415 [M + H]+.
d) 4-Hydroxy- 1 -( 1 -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)pyridin-2( lH)-one
Chemical Formula: CIgH2ON4O2
Exact Mass: 324.1586 Molecular Weight: 324.377
Figure imgf000046_0002
[0081] To a solution of 4-(benzyloxy)-l-(l-(2-(pyrrolidin-l-yl)ethyl)-lH-indazol-6- yl)pyridin-2(lH)-one (240 mg, 0.58 mmol) in CH3OH was added Pd/C (200 mg) under an Ar atmosphere. The Ar balloon was replaced with a H2 balloon. The reaction was heated to 55°C overnight and then allowed to cool. The mixture was filtered through a layer of Celite and the filtrate was concentrated. Purification by flash column chromatography (silica gel, CH2Cl2/Me0H, 80:20) gave the title compound as a white solid in 65% yield: 1H NMR (500 MHz, CD3OD) δ 8.12 (s, IH), 7.74 (d, J = 2.0 Hz, IH), 7.71 (d, J = 9.0 Hz, IH), 7.48 (d, J= 7.5 Hz, IH), 7.39 (dd, J= 9.0, 2.0 Hz, IH), 6.12 (d, J= 7.5 Hz, IH), 4.65 (t, J= 7.0 Hz, 2H), 3.17 (t, J = 7.0 Hz, 2H), 2.72-2.71 (m, 4H), 1.84-1.80 (m, 4H); ESI MS m/z 325 [M + H]+.
e) 1 -(2-(Pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)pyridin-2(lH)-one-4- trifluoromethanesulfonate
Figure imgf000047_0001
[0082] 4-Hydroxy- 1 -( 1 -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)pyridin-2( lH)-one (900 mg, 2.7 mmol) was suspended in THF (20 niL) under a nitrogen atmosphere and LiN(SiMe3)2 (IM in THF) (4.2 niL, 4.2 mmol) added. After stirring for 1 minute, PhNTf2 (1.48 g, 4.16 mmol) was added in one portion and the mixture was stirred for 2 h. The mixture was concentrated, diluted with methylene chloride (50 mL) and washed successively with saturated NH4Cl and saturated Na2CO3, dried over sodium sulfate and concentrated. The residue was purified by column chromatography (40 g ISCO column eluting with methylene chloride and a methanol/ammonia mixture (10:1); gradient 100% methylene chloride to 80% methylene chloride over 30 min at 40 mL/min) to provide the title compound (780 mg, 60%) as a white solid; 1H NMR (500 MHz, CDCl3) δ 8.05 (s, IH), 7.68 (d, J = 1.8 Hz, IH), 7.57 (d, J = 8.9 Hz, IH), 7.53 (d, J = 7.4 Hz, IH), 7.37-7.35 (dd, J = 8.9, 2.0 Hz, IH), 6.60 (d, J = 2.7 Hz, IH), 6.30-6.28 (dd, J = 8.7, 2.5 Hz, IH), 4.57 (t, J = 7.2 Hz, 2H), 3.01 (t, J = 7.2 Hz, 2H), 2.60-2.57 (m, 4H), 1.80-1.76 (m, 4H) ; ESI MS m/z 457 [M + H]+.
f) 4-(PhenylV 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol-5-yr)pyridin-2( lHVone hydrochloride
Figure imgf000048_0001
[0083] 1 -(2-(Pyrrolidin- 1 -yl)ethyl)- l/f-indazol-5-yl)pyridin-2(lH)-one-4-trifluorometh- anesulfonate (100 mg, 0.22 mmol), phenylboronic acid (67 mg, 0.55 mmol), K2CO3 (75 mg, 0.55 mmol) and [l,r-Bis-(diphosphenylphosphino)ferrocene]dichloropalladium(II) (PdCl2dppf) (18 mg, 0.022 mmol) were stirred in DMSO (2 mL) under vacuum for 30 min. The flask was flushed with nitrogen and the mixture was heated to 8O0C for 10 min. Upon cooling, the mixture was diluted with methylene chloride and washed with 5% lithium chloride solution (5x), dried, concentrated and the residue was purified by column chromatography (12 g ISCO column eluting with methylene chloride and a methanol/ammonia mixture (10:1); gradient 100% methylene chloride to 80% methylene chloride over 30 min at 25 mL/min) to provide the free base. This was dissolved in methylene chloride (2 mL) and treated with 1 equivalent of 2 M HCl in Et2O and the mixture was concentrated to provide the title compound (20 mg, 21%) as a yellow solid: melting point (mp) 192-196°C; 1H NMR (500 MHz, CD3OD) δ 8.27 (s, IH), 7.93 (d, J = 1.7 Hz, IH), 7.86-7.82 (m, 2H), 7.79-7.76 (m, 2H), 7.57-7.52 (m, 4H), 6.97-6.96 (m, 2H), 4.90 (t, J = 5.7 Hz, 2H), 3.88 (t, J = 5.7 Hz, 2H), 3.74-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.20-2.14 (m, 2H), 2.06-2.00 (m, 2H); ESI MS m/z 385 [M + H]+; HPLC (Method D) >99% (AUC), & = 13.0 min.
Example 2
Preparation of 4-(4-ChlorophenyD- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- l/-f-indazol-5-yl)pyridin- 2(l/-/)-one hydrochloride
Figure imgf000049_0001
[0084] Following the procedure of Example 1, but substituting /?-chlorophenylboronic acid for phenylboronic acid, the title compound (26 mg, 24%) was prepared as a yellow solid: mp 255-2600C (decomposition); 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J = 1.6 Hz, IH), 7.86-7.82 (m, 2H), 7.77 (d, J= 8.6 Hz, 2H), 7.56-7.53 (m, 3H), 6.95-6.92 (m, 2H), 4.90 (t, J = 5.6 Hz, 2H), 3.87 (t, J = 5.6 Hz, 2H), 3.73-3.69 (m, 2H), 3.19-3.14 (m, 2H), 2.18-2.15 (m, 2H), 2.03-2.00 (m, 2H); ESI MS m/z 419 [M + H]+; HPLC (Method D) >99% (AUC), tR = 13.9 min.
Example 3
Preparation of 4-(Benzo[^lthiophen-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Figure imgf000049_0002
[0085] Following the procedure of Example 1, but substituting benzothiophene-2-boronic acid for phenylboronic acid, the title compound (23 mg, 22%) was prepared as a yellow solid: mp 260-2640C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 8.03 (s, IH), 7.95-7.91 (m, 3H), 7.83 (d, J = 8.9 Hz, IH), 7.77 (d, J = 7.1 Hz, IH), 7.56-7.54 (dd, J = 8.9, 1.9 Hz, IH), 7.46-7.41 (m, 2H), 7.06-7.02 (dd, J = 7.2, 2.0 Hz, IH), 6.97 (d, J = 1.8 Hz, IH), 4.89 (t, J = 5.8 Hz, 2H), 3.87 (t, J = 5.8 Hz, 2H), 3.73-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.19- 2.16 (m, 2H), 2.04-2.00 (m, 2H); ESI MS m/z 441 [M + H]+; HPLC (Method D) >99% (AUC), tR = 14.5 min.
Example 4
Preparation of 4-(Benzo furan-2-yl)- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- lH-indazo 1-5 -yDpyridin- 2(l/-/)-one hydrochloride
Figure imgf000050_0001
[0086] Following the procedure of Example 1, but substituting benzofuran-2-boronic acid for phenylboronic acid, the title compound (21 mg, 21%) was prepared as a brown solid: mp 256-2600C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J= 1.7 Hz, IH), 7.85 (d, J = 8.9 Hz, IH), 7.78 (d, J = 7.1 Hz, IH), 7.71 (d, J = 7.8 Hz, IH), 7.61 (d, J = 8.3 Hz, IH), 7.60 (s, IH), 7.56-7.53 (dd, J = 8.9, 1.9 Hz, IH), 7.45-7.41 (t, J = 8.3 Hz, IH), 7.31 (t, J = 7.8 Hz, IH), 7.15 (d, J = 1.8 Hz, IH), 7.05-7.03 (dd, J = 7.1, 1.8 Hz, IH) 4.91 (t, J = 5.3 Hz, 2H), 3.88 (t, J= 5.3 Hz, 2H), 3.73-3.69 (m, 2H), 3.20-3.14 (m, 2H), 2.18-2.14 (m, 2H), 2.04-1.99 (m, 2H); ESI MS m/z 425 [M + H]+; HPLC (Method E) 99% (AUC), tR = 15.4 min.
Example 5
Preparation of 4-(4-FluorophenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- lH-indazo 1-5 -yPpyridin- 2(l/-/)-one hydrochloride
Figure imgf000051_0001
[0087] Following the procedure of Example 1, but substituting /7-fluorophenylboronic acid for phenylboronic acid, the title compound (18 mg, 19%) was prepared as a yellow solid: mp 115-1200C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J = 1.4 Hz, IH), 7.83- 7.79 (m, 3H), 7.77 (d, J = 7.1 Hz, IH), 7.55-7.53 (dd, J = 8.8, 1.9 Hz, IH), 7.26-7.25 (m, 2H), 6.89 (d, J = 1.8 Hz, IH), 6.87-6.85 (dd, J = 7.2, 2.1 Hz, IH), 4.87 (t, J = 5.7 Hz, 2H), 3.87 (t, J = 5.7 Hz, 2H), 3.76-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.19-2.16 (m, 2H), 2.03- 2.00 (m, 2H); ESI MS m/z 403 [M + H]+; HPLC (Method D) >99% (AUC), tR = 13.2 min.
Example 6
Preparation of 4-(Naphthalen-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- l/-f-indazol-5-yl)pyridin- 2(l/-/)-one hydrochloride
Figure imgf000051_0002
[0088] Following the procedure of Example 1, but substituting napthyl-2-boronic acid for phenylboronic acid, the title compound (24 mg, 24%) was prepared as a yellow solid: mp 128-134°C; 1H NMR (500 MHz, CD3OD) δ 8.32 (s, IH), 8.27 (s, IH), 8.04-8.01 (m, 2H), 7.95-7.93 (m, 2H), 7.87-7.81 (m, 3H), 7.59-7.56 (m, 3H), 7.07-7.05 (m, 2H), 4.89 (t, J = 5.8 Hz, 2H), 3.88 (t, J = 5.8 Hz, 2H), 3.74-3.70 (m, 2H), 3.19-3.16 (m, 2H), 2.19-2.16 (m, 2H), 2.04-2.01 (m, 2H); ESI MS m/z 435 [M + H]+; HPLC (Method D) >99% (AUC), tR = 14.4 min.
Example 7
Preparation of 4-(3-Chlorophenyl)- 1 -(I -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol-5-yDpyridin- 2(lH)-one hydrochloride
Figure imgf000052_0001
[0089] Following the procedure of Example 1, but substituting m-chlorophenylboronic acid for phenylboronic acid, the title compound (20 mg, 21%) was prepared as an orange solid: mp 118-125°C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J= 1.6 Hz, IH), 7.83 (d, J = 8.9 Hz, IH), 7.79-7.78 (m, 2H), 7.70-7.68 (m, IH), 7.55-7.52 (m, 3H), 6.91 (d, J =1.8 Hz, IH), 6.86-6.84 (dd, J = 7.1, 2.0 Hz, IH), 4.90 (t, J = 5.8 Hz, 2H), 3.88-3.86 (m, 2H), 3.76-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.19-2.16 (m, 2H), 2.03-2.00 (m, 2H); ESI MS m/z 419 [M + H]+; HPLC (Method D) 98.5% (AUC), tR = 13.9 min.
Example 8
Preparation of 4-(3-PhenoxyphenyD- 1 -(I -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol-5- yl)pyridin-2(l//)-one hydrochloride
Figure imgf000052_0002
[0090] Following the procedure of Example 1, but substituting 3-phenoxyphenylboronic acid for phenylboronic acid, the title compound (18 mg, 16%) was prepared as a yellow- brown solid: 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.90 (d, J= 1.7 Hz, IH), 7.82 (d, J= 8.9 Hz, IH), 7.76 (d, J= 7.1 IH), 7.54-7.49 (m, 3H), 7.41-7.38 (m, 2H), 7.36-7.35 (m, IH), 7.16 (t, J = 8.5 Hz, IH), 7.12-7.10 (dt, J = 6.7, 2.4 Hz, IH), 7.07-7.05 (m, 2H), 6.86 (d, J= 1.8 Hz, IH), 6.83-6.81 (dd, J= 7.3, 2.0 Hz, IH), 4.89 (t, J= 5.7 Hz, 2H), 3.87 (t, J = 5.7 Hz, 2H), 3.73-3.69 (m, 2H), 3.20-3.14 (m, 2H), 2.18-2.15 (m, 2H), 2.04-2.00 (m, 2H); ESI MS m/z 477 [M + H]+; HPLC (Method D) >99% (AUC), tR = 15.3 min.
Example 9
Preparation of 1 -(I -(2-(Pyrroridin- 1 -vOethvO- lH-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
Figure imgf000053_0001
[0091] Following the procedure of Example 1, but substituting p- trifluoromethylphenylboronic acid for phenylboronic acid, the title compound (35 mg, 31%) was prepared as a white solid: 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.96 (d, J= 8.2 Hz, 2H), 7.92 (d, J = 1.8 Hz, IH), 7.85-7.83 (m, 3H), 7.82 (d, J = 7.2 Hz, IH), 7.56-7.54 (dd, J= 8.9, 1.9 Hz, IH), 6.97 (d, J= 1.9 Hz, IH), 6.89-6.87 (dd, J= 7.1, 2.0 Hz, IH), 4.90 (t, J = 5.7 Hz, 2H), 3.88 (t, J = 5.7 Hz, 2H), 3.74-3.69 (m, 2H), 3.20-3.16 (m, 2H); 2.19- 2.16 (m, 2H), 2.04-2.00 (m, 2H); ESI MS m/z 453 [M + H]+; HPLC (Method E) >99% (AUC), to = 13.4 min.
Example 10
Preparation of 1 -(I -(2-(Pyrroridin- 1 -vOethvO- lH-indazol-5-yl)-4-(3- (trifluoromethyl)phenyl)pyridin-2( lHVone dihydrochloride
Figure imgf000054_0001
[0092] Following the procedure of Example 1, but substituting m- trifluoromethylphenylboronic acid for phenylboronic acid, the title compound (46 mg, 40%) was prepared as a brown solid: 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 8.08-7.99 (br m, 2H), 7.92 (d, J= 1.5 Hz, IH), 7.85-7.81 (m, 3H), 7.75 (t, J= 8.0 Hz, IH), 7.56-7.44 (dd, J= 8.9, 1.9 Hz, IH), 6.96 (d, J= 1.8 Hz, IH), 6.90-6.89 (dd, J= 7.1, 1.9 Hz, IH), 4.90 (t, J = 5.8 Hz, 2H), 3.88 (t, J = 5.8 Hz, 2H), 3.73-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.20-2.13 (m, 2H), 2.05-2.00 (m, 2H); ESI MS m/z 453 [M + H]+; HPLC (Method C) 96.9% (AUC), tκ = 13.3 min.
Example 11
Preparation of 4-( 1 -Methyl- lH-indo 1-2- vD- 1 -( 1 -(2-(pyrrolidin- 1 - vOethvO- lH-indazo 1-5 - yl)pyridin-2( 1 H)-one dihydrochloride
Figure imgf000054_0002
[0093] Following the procedure of Example 1, but substituting JV-methylindole-2-boronic acid for phenylboronic acid, the title compound (27 mg, 24%) was prepared as a brown solid: 1H NMR (500 MHz, DMSO-J6) δ 10.71-10.48 (br s, IH), 8.28 (s, IH), 7.94 (d, J = 6.2 Hz, IH), 7.92 (s, IH), 7.82 (d, J = 7.2 Hz, IH), 7.63 (d, J = 7.8 Hz, IH), 7.57 (m, 2H), 7.27 (d, J = 8.1 Hz, IH), 7.11 (t, J = 7.0 Hz, IH), 6.88 (s, IH), 6.71 (d, J = 1.8 Hz, IH), 6.66-6.63 (dd, J = 7.1, 1.9 Hz, IH), 4.91 (t, J = 6.3 Hz, 2H), 3.89 (s, 3H), 3.74 (q, J = 6.2 Hz, 2H), 3.53-3.50 (m, 2H), 3.08-3.00 (m, 2H) 1.99-1.83 (m, 4H); ESI MS m/z 438 [M + H]+; HPLC (Method C) >99% (AUC), tR = 13.2 min.
Example 12
Preparation of 4-(2,4-DichlorophenyD- 1 -(I -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Figure imgf000055_0001
[0094] Following the procedure of Example 1, but substituting 2,4-dichlorophenylboronic acid for phenylboronic acid, the title compound (29 mg, 27%) was prepared as a white solid: mp 110-1150C deliquescent; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.92 (d, J = 1.5 Hz, IH), 7.82 (d, J= 8.9 Hz, IH), 7.76 (d, J= 7.0 Hz, IH), 7.65 (s, IH), 7.56-7.54 (dd, J = 8.8, 1.8 Hz, IH), 7.49 (s, 2H), 6.69 (d, J= 1.5 Hz, IH), 6.22-6.20 (dd, J= 7.0, 1.8 Hz, IH), 4.90 (t, J = 5.7 Hz, 2H), 3.87 (t, J = 5.7 Hz, 2H), 3.73-3.69 (br m, 2H), 3.20-3.15 (br m, 2H), 2.19-2.16 (m, 2H), 2.03-2.00 (m, 2H); ESI MS m/z 453 [M + H]+; HPLC (Method D) >99% (AUC), tR = 14.5 min.
Example 13
Preparation of 4-(Naphthalen- 1 -yl)- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- lH-indazo 1-5 -yl) pyridin- 2(l/-/)-one dihydrochloride:
Figure imgf000056_0001
[0095] Following the procedure of Example 1, but substituting napthyl-1-boronic acid for phenylboronic acid, the title compound (30.5 mg, 22%) was prepared as orange crystals: mp 128-133°C; 1H NMR (500 MHz, CDCl3 + CD3OD) δ 8.22 (s, IH), 8.05-8.03 (m, IH), 7.99- 7.96 (m, 2H), 7.92 (s, 2H), 7.68 (d, J= 6.8 Hz, IH), 7.62-7.54 (m, 5H), 6.86 (s, IH), 6.70- 6.68 (dd, J= 5.4, 1.4 Hz, IH), 4.97-4.96 (br m, 2H), 3.86 (br m, 2H), 3.67-3.66 (br m, 2H), 3.06-2.96 (br m, 2H), 2.13-2.08 (br m, 4H); ESI MS m/z 435 [M + H]+. HPLC (Method C) 98.7% (AUC), tR = 13.3 min.
Example 14
Preparation of 1 -( 1 -(2-(Pyrrolidin- 1 -yDethyl)- l/-f-indazol-5-yl)-4-p-tolylpyridin-2( lHVone hydrochloride
Figure imgf000056_0002
[0096] Following the procedure of Example 1, but substituting /?-tolylboronic acid for phenylboronic acid, the title compound (52 mg, 55%) was prepared as a brown solid: mp 240-2460C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.89 (d, J= 1.5 Hz, IH), 7.81 (d, J = 8.9 Hz, IH), 7.32 (d, J= 7.3 Hz, IH), 7.65 (d, J= 8.2 Hz, 2H), 7.54-7.52 (dd, J= 8.9, 2.0 Hz, IH), 7.34 (d, J = 8.0 Hz, 2H), 6.88 (d, J = 1.6 Hz, IH), 6.86-6.84 (dd, J = 7.1, 2.0 Hz, IH), 4.86 (t, J = 5.8 Hz, 2H), 3.81 (t, J = 5.8 Hz, 2H), 3.43-3.42 (br m, 4H), 2.41 (s, 3H), 2.12-1.99 (m, 4H); ESI MS m/z 399 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.7 min.
Example 15
Preparation of 1 -(I -(2-(Pyrrolidin- 1 -vOethvO- lH-indazol-5-yl)-4-(4- (trifluoromethoxy)phenyl)pyridin-2( lHVone hydrochloride
Figure imgf000057_0001
[0097] Following the procedure of Example 1, but substituting p- trifluoromethoxyphenylboronic acid for phenylboronic acid, the title compound (51 mg, 46%) was prepared as an orange solid: mp 202-2090C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J= 1.6 Hz, IH), 7.88 (d, J= 8.7 Hz, 2H), 7.84 (d, J= 8.9 Hz, IH), 7.80 (d, J = 7.1 Hz, IH), 7.55-7.53 (dd, J = 8.7 Hz, IH), 7.45 (d, J = 8.3 Hz, 2H), 6.93 (d, J = 1.7 Hz, IH), 6.88-6.86 (dd, J = 7.2, 2.0 Hz, IH), 4.89 (t, J = 6.8 Hz, 2H), 3.89 (t, J = 6.8 Hz, 2H), 3.74-3.69 (br m, 2H), 3.20-3.15 (br m, 2H), 2.18-2.15 (m, 2H), 2.03-2.00 (m, 2H); ESI MS m/z 469 [M + H]+; HPLC (Method C) >99% (AUC), to = 13.7 min.
Example 16
Preparation of 4-0 -Methyl- lH-indo 1-5 - vD- 1 -( 1 -(2-(pyrrolidin- 1 - vOethvO- lH-indazo 1-5 - yl)pyridin-2Q/-/)-one hydrochloride
Figure imgf000058_0001
[0098] Following the procedure of Example 1, but substituting JV-methylindole-5-boronic acid for phenylboronic acid, the title compound (36 mg, 37%) was prepared as a brown solid: 1H NMR (500 MHz, CD3OD) 5 8.26 (s, IH), 8.01 (d, J= 1.5 Hz, IH), 7.91 (d, J= 1.5 Hz, IH), 7.81 (d, J = 8.9 Hz, IH), 7.71 (d, J = 7.1 Hz, IH), 7.60-7.58 (dd, J = 8.6, 1.8 Hz, IH), 7.56-7.54 (dd, J= 8.9, 1.9 Hz, IH), 7.52 (d, J= 8.7 Hz, IH), 7.26 (d, J= 3.1 Hz, IH), 6.97 (dd, J= 7.1, 2.0 Hz, IH), 6.94 (d, J= 1.8 Hz, IH), 6.57 (d, J= 3.2 Hz, IH), 4.88 (t, J = 5.7 Hz, 2H), 3.87-3.85 (s overlapping with m, 5H), 3.80-3.10 (br m, 4H), 2.09 (br m, 4H), ESI MS m/z 438 [M + H]+; HPLC (Method C) 98.4% (AUC), tR = 12.6 min.
Example 17
Preparation of 4-(4-CyanophenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazo 1-5 -yPpyridin- 2(l/-/)-one hydrochloride
Figure imgf000058_0002
[0099] Following the procedure of Example 1, but substituting /?-cyanophenylboronic acid for phenylboronic acid, the title compound (23 mg, 23%) was prepared as a yellow solid: mp 92-96°C; 1H NMR (500 MHz, CD3OD) δ 8.23 (s, IH), 7.94 (d, J = 6.6 Hz, 2H), 7.90-7.88 (m, 3H), 7.82 (d, J= 2.8 Hz, IH), 7.80 (s, IH), 7.53-7.51 (dd, J= 8.9,1.9 Hz, IH), 6.95 (d, J = 1.9 Hz, IH), 6.86-6.84 (dd, J = 7.1, 2.1 Hz, IH), 4.82 (m, 2H), 3.66 (br m, 2H), 3.26 (br m, 4H), 2.01 (br m, 4H); ESI MS m/z 410 [M + H]+; HPLC (Method C) >99% (AUC), tR = 11.6 min.
Example 18
Preparation of 4-(4-Methoxyphenyl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Figure imgf000059_0001
[00100] Following the procedure of Example 1, but substituting /?-methoxyphenylboronic acid for phenylboronic acid, the title compound (45 mg, 46%) was prepared as an orange solid: mp 105-1100C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J= 1.5 Hz, IH), 7.83 (d, J= 6.9 Hz, IH), 7.78-7.43 (m, 3H), 7.56-7.53 (dd, J= 8.9 Hz, IH), 7.09 (d, J= 8.9 Hz, 2H), 6.96-6.94 (dd, J = 7.1, 2.2 Hz, IH), 6.93 (d, J = 1.7 Hz, IH), 4.88 (t, J = 5.4 Hz, 2H), 3.88-3.86 (s overlapping with m, 5H), 3.73-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.19- 2.14 (m, 2H), 2.03-2.06 (m, 2H); ESI MS m/z 415 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.0 min.
Example 19
Preparation of 4-(4-Fluoro-2-cyanophenyD- 1 -(I -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol-5- yl)pyridin-2(l/-/)-one hydrochloride
Figure imgf000060_0001
[00101] Following the procedure of Example 1, but substituting 2-cyano-4- fluorophenylboronic acid pinacol ester for phenylboronic acid, the title compound (45 mg, 44%) was prepared as a brown solid: mp 81-86 0C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.94-7.93 (m, IH), 7.83 (d, J = 2.1 Hz, IH), 7.82 (s, IH), 7.77-7.74 (m, 2H), 7.63- 7.59 (dt, J= 8.3, 2.7 Hz, IH), 7.57-7.53 (dd, J= 8.9, 2.0 Hz, IH), 6.83 (d, J= 1.9 Hz, IH), 6.73-6.70 (dd, J = 7.0, 2.0 Hz, IH), 4.89 (t, J = 5.8 Hz, 2H), 3.89-3.86 (t, J = 5.8 Hz, 2H), 3.72-3.61 (br m, 2H), 3.26-3.15 (br m, 2H), 2.11-2.05 (m, 4H); ESI MS m/z 428 [M + H]+; HPLC (Method C) 98.5% (AUC), tR = 11.7 min.
Example 20
Preparation of 4-(2,4-DimethoxyphenyD- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- l/-f-indazol-5-yl) pyridin-2(l/-/)-one hydrochloride:
Figure imgf000060_0002
[00102] Following the procedure of Example 1, but substituting 2,4- dimethoxyphenylboronic acid for phenylboronic acid, the title compound (29.7 mg, 61%) was prepared as orange crystals: mp 95-115°C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.93 (d, J = 1.7 Hz, IH), 7.84 (d, J = 8.9 Hz, IH), 7.73 (d, J= 8.1 Hz, IH), 7.56-7.54 (dd, J = 8.9, 2.0 Hz, IH), 7.45 (d, J = 8.4 Hz, IH) 6.96-6.92 (m, 2H), 6.70-6.67 (m, 2H), 4.89 (t, J = 5.8 Hz, 2H), 3.89-3.87 (m, 8H), 3.73-3.69 (br m, 2H), 3.19-3.16 (br m, 2H) 2.17-2.16 (br m, 2H) 2.03-2.01 (br m, 2H); ESI MS m/z 445 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.2 min.
Example 21
Preparation of 4-(2,4-DifluorophenyD- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- l/-f-indazol-5-yl) pyridin-2(lH)-one hydrochloride:
Figure imgf000061_0001
[00103] Following the procedure of Example 1, but substituting 2,4-difluorophenylboronic acid for phenylboronic acid, the title compound (20.4 mg, 45%) was prepared as a brown solid: mp 235-255°C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.91 (d, J= 1.4 Hz, IH), 7.80 (d, J= 8.9 Hz, IH), 7.75 (d, J= 6.9 Hz, IH), 7.71-7.62 (m, IH), 7.55-7.53 (dd, J= 8.8, 1.9 Hz, IH), 7.18-7.12 (m, 2H), 6.82 (s, IH), 6.74-6.71 (dt, J= 7.1, 1.9 Hz, IH), 4.88 (t, J = 5.8, 2H), 3.85 (t, J= 5.7 Hz, 2H), 3.56-3.32 (br m, 4H), 2.20-1.91 (br m, 4H); ESI MS m/z 421 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.3 min.
Example 22
Preparation of 4-(4-Isopropoxyphenyl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- l/-f-indazol-5-yl) pyridine-2(l/-/)-one hydrochloride:
Figure imgf000062_0001
[00104] Following the procedure of Example 1, but substituting 4- ώopropoxyphenylboronic acid for phenylboronic acid, the title compound (63 mg, 65%) was prepared as gray crystals: mp 235-2500C; 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.89 (d, J = 1.4 Hz, IH), 7.81 (d, J = 8.9 Hz, IH), 7.72-7.69 (m, 3H), 7.54-7.51 (dd, J = 8.9, 2.0 Hz, IH), 7.05-7.02 (d, J = 8.8 Hz, 2H), 6.86-6.84 (m, 2H), 4.87 (t, J = 5.8 Hz, 2H), 4.72- 4.67 (m, IH), 3.84 (t, J= 5.7 Hz, 2H), 3.50-3.32 (br m, 4H), 2.15-2.01 (br m, 4H), 1.35 (d, J = 6.0 Hz, 6H); ESI MS m/z 443 [M + H]+; HPLC (Method C) 98.3% (AUC), tR = 13.2 min.
Example 23
Preparation of 4-(2,4-to(Trifluoromethyl)phenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- IH- indazo 1-5 - yl)pyridin-2( lH)-one hydrochloride :
Figure imgf000062_0002
[00105] Following the procedure of Example 1, but substituting 2,4- δώ(trifluoromethyl)phenylboronic acid for phenylboronic acid, the title compound (24.9 mg, 5%) was prepared as gray crystals: mp 235-245°C; 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 8.13 (s, IH) 8.09 (d, J = 8.0 Hz, IH), 7.93 (d, J = 1.7, IH), 7.82 (d, J = 9.0 Hz, IH), 7.78 (d, J= 7.0 Hz, IH), 7.75 (d, J= 8.1 Hz, IH), 7.56-7.54 (dd, J= 8.9, 1.9 Hz, IH), 6.64
(d, J= 1.7 Hz, IH), 6.53-6.51 (dd, J= 7.0, 1.8 Hz, IH), 4.87-4.84 (br m, 2H), 3.81-3.68 (br m, 2H), 3.44-3.34 (br m, 4H), 2.12-1.94 (br m, 4H); ESI MS m/z 521 [M + H]+; HPLC (Method C) >99% (AUC), tR = 14.2 min.
Example 24
Preparation of 4-(4-Butoxy-2-methylphenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazo 1-5 - yl)pyridin-2(lH)-one hydrochloride:
Figure imgf000063_0001
[00106] Following the procedure of Example 1, but substituting 4-(butyloxy)-2- methylphenylboronic acid for phenylboronic acid, the title compound (82.3 mg, 24%) was prepared as brown crystals: mp 80-950C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.92 (d, J = 1.4 Hz, IH), 7.82 (d, J = 8.9 Hz, IH), 7.70 (d, J = 6.9 Hz, IH), 7.56-7.54 (dd, J = 8.9, 2.0 Hz, IH), 7.24 (d, J = 8.4 Hz, IH), 6.88 (d, J = 2.4 Hz, IH), 6.86-6.84 (dd, J = 8.4, 2.6, IH), 6.58-6.56 (m, 2H), 4.89 (t, J = 5.8 Hz, 2H), 4.02 (t, J = 6.7 Hz, 2H), 3.87 (t, J = 5.8 Hz, 2H), 3.74-3.69 (br m, 2H), 3.20-3.15 (br m, 2H), 2.39 (s, 3H), 2.19-2.16 (br m, 2H), 2.04-2.00 (br m, 2H), 1.80-1.75 (m, 2H), 1.55-1.49 (m, 2H), 1.00 (t, J = 7.4 Hz, 3H); ESI MS m/z 471 [M + H]+; HPLC (Method C) 94.6% (AUC), tR = 14.8 min.
Example 25
Preparation of 1 -( 1 -(2-(Pyrrolidin- 1 -yPethyl)- lH-indazol-5-yl)-4-o-tolylpyridin-2( lHVone hydrochloride:
Figure imgf000064_0001
[00107] Following the procedure of Example 1, but substituting o-tolylboronic acid for phenylboronic acid, the title compound (34.3 mg, 79%) was prepared as orange crystals: mp 80-950C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.93 (d, J= 1.5 Hz, IH) 7.82 (d, J = 8.9 Hz, IH), 7.72 (d, J = 7.0 Hz, IH), 7.57 (dd, J = 8.9, 2.1 Hz, IH), 7.35-7.33 (m, 2H), 7.30-7.29 (m, 2H), 6.58 (d, J= 1.4 Hz, IH), 6.55-6.53 (dd, J= 7.0, 2.2 Hz, IH), 4.89 (t, J = 5.7 Hz, 2H), 3.87 (t, J = 5.7 Hz, 2H), 3.76-3.67 (br m, 2H), 3.19-3.15 (br m, 2H), 2.39 (s, 3H) 2.22-2.11 (br m, 2H), 2.03-2.01 (br m, 2H); ESI MS m/z 399 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.4 min.
Example 26
Preparation of 4-(4-(Benzyloxy)-2-methylphenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- IH- indazo 1-5 - yl)pyridin-2( lH)-one hydrochloride :
Figure imgf000064_0002
[00108] Following the procedure of Example 1, but substituting 4-(benzyloxy)-2- methylphenylboronic acid for phenylboronic acid, the title compound (45.5 mg, 51%) was prepared as orange crystals: mp 75-85°C; 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.91 (d, J = 1.8 Hz, IH), 7.81 (d, J = 8.9 Hz, IH), 7.69 (d, J = 6.9 Hz, IH), 7.55-7.53 (dd, J = 8.9, 1.9 Hz, IH), 7.45 (d, J= 7.3 Hz, 2H), 7.38 (t, J= 7.5 Hz 2H), 7.32 (d, J= 5.3, IH), 7.25 (d, J= 8.5 Hz, IH), 6.98 (d, J= 2.5 Hz, IH), 6.95-6.93 (dd, J= 8.4, 2.6 Hz, IH), 6.56 (d, J = 1.7 Hz, IH), 6.55-6.53 (dd, J = 6.9, 1.9 Hz, IH), 5.13 (s, 2H), 4.87 (t, J = 5.8 Hz, 2H), 3.88-3.79 (br m, 2H), 3.34-3.32 (br m, 4H), 2.38 (s, 3H) 2.16-2.00 (br m, 4H); ESI MS m/z 505 [M + H]+. HPLC (Method C) >99% (AUC), tR = 14.7 min.
Example 27
Preparation of 4-(4-Chloro-2-methoxyphenyl)- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride:
Figure imgf000065_0001
[00109] Following the procedure of Example 1, but substituting 4-chloro-2- methoxyphenylboronic acid for phenylboronic acid, the title compound (22.1 mg, 30%) was prepared as a white powder: mp 248-256°C; 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.90 (d, J= 1.4 Hz, IH), 7.81 (d, J= 8.9 Hz, IH), 7.66 (d, J= 7.1 Hz, IH), 7.54-7.52 (dd, J = 8.9, 1.9 Hz, IH), 7.42 (d, J= 8.2 Hz, IH) 7.19 (d, J= 1.8 Hz, IH), 7.11-7.09 (dd, J= 8.2, 1.9 Hz, IH), 6.78 (t, J= 1.5 Hz 2H), 4.88 (t, J= 5.6 Hz, 2H), 3.89 (s, 3H ), 3.85 (t, J= 5.7, 2H), 3.80-3.33 (br m, 4H), 2.17-2.00 (br m, 4H); ESI MS m/z 449 [M + H]+; HPLC (Method C) 99.9% (AUC), tR = 13.1 min.
Example 28
Preparation of 4-(Benzo[άH [1 ,3]dioxol-5-yl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyP)- lH-indazol-5- yl)pyridin-2(l/-/)-one hydrochloride
Figure imgf000066_0001
[00110] Following the procedure of Example 1, but substituting 3,4- methylenedioxyphenylboronic acid for phenylboronic acid, the title compound (24.1 mg, 52%) was prepared as orange-brown crystals: mp 75-85°C; 1U NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.90 (d, J = 1.4 Hz, IH) 7.82 (d, J = 8.9 Hz, IH), 7.73-7.71 (dd, J = 5.1, 2.7 Hz, IH), 7.54-7.52 (d, J = 7.9, IH), 7.32-7.30 (dd, J = 8.1, 1.9 Hz, IH), 7.27 (d, J = 1.8, IH), 6.97 (d, J= 8.1 Hz, IH), 6.85-6.83 (m, 2H), 6.05 (s, 2H), 4.88 (t, J= 5.8 Hz, 2H), 3.87 (t, J = 5.8 Hz, 2H), 3.74-3.69 (br m, 2H), 3.20-3.15 (br m, 2H) 2.19-2.16 (br m, 2H), 2.03- 2.00 (br m, 2H); ESI MS m/z 429 [M + H]+; HPLC (Method C) 97.7% (AUC), to = 11.7 min.
Example 29
Preparation of 4-(4-Methoxy-2-methylphenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol 5-yl)pyridin-2(lH)-one hydrochloride
Figure imgf000066_0002
[00111] Following the procedure of Example 1, but substituting 4-methxoy-2- methylphenylboronic acid for phenylboronic acid, the title compound (10.3 mg, 20%) was prepared as orange-brown crystals: mp 205-2150C; 1U NMR (500 MHz, CD3OD) δ 8.26 (s,
IH), 7.91 (s, IH), 7.82 (d, J = 8.6 Hz, IH), 7.69 (d, J = 6.9 Hz, IH), 7.55 (d, J = 8.5 Hz, IH), 7.25 (d, J= 8.5 Hz, IH), 6.90-6.86 (m, 2H), 6.56-6.53 (m, 2H), 4.92-4.85 (br m, 2H), 3.90-3.79 (br m, 5H), 3.75-3.32 (br m, 4H), 2.39 (s, 3H), 2.16-2.01 (br m, 4H); ESI MS m/z 429 [M + H]+; HPLC (Method D) 98.6% (AUC), tR = 14.2 min.
Example 30
Preparation of 4-(5 -Methylpyridin-2-yl)- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazo 1-5 - yl)pyridin-2(lH)-one hydrochloride
a) 1 -( 1 -(2-(Pyrrolidin- 1 -yl)ethyl)- l/f-indazol-5-yl)-4-(trimethylstannyl)pyridin-2( lH)-one
Figure imgf000067_0001
[00112] 1 -(2-(Pyrrolidin- 1 -yl)ethyl)- l/f-indazol-5-yl)pyridin-2(lH)-one-4- trifluoromethanesulfonate (250 mg, 0.54 mmol) and hexamethylditin (360 mg, 1.1 mmol) were stirred in dry toluene (5 mL)/DMSO (1 mL) and degassed with a nitrogen stream as the temperature was increased to 1000C. Palladium tetrakistriphenylphosphine (62 mg, 0.054 mmol) was added and the reaction was maintained at 1000C under a nitrogen atmosphere for 2 h. Upon cooling, the mixture was purified by Combiflash chromatography (12 g ISCO column eluting with methylene chloride and methanol/ammonia (10:1); 100% methylene chloride to 20% methanol/ammonia over 30 min at 25 mL/min). Concentration of the appropriate fractions provided the desired stannane (143 mg, 56%) as a colorless oil; 1H NMR (500 MHz, CDCl3) δ 8.03 (s, IH), 7.67 (d, J = 1.5 Hz, IH), 7.52 (d, J = 8.9 Hz, IH), 7.41-7.39 (dd, J= 8.9, 1.0 Hz, IH), 7.29 (d, J= 6.6 Hz, IH), 6.84 (s, IH), 6.32-6.30 (dd, J = 6.6, 0.9 Hz, IH), 4.55 (t, J = 7.3 Hz, 2H), 3.00 (t, J = 7.3 Hz, 2H), 2.60-2.57 (m, 4H), 1.80-1.77 (m, 4H), 0.34 (s, 9H).
b) 4-(5 -Methylpyridin-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)pyridin-2(lH)- one hydrochloride
Figure imgf000068_0001
[00113] 1 -(I -(2-(Pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)-4-(trimethylstannyl)pyridin- 2(lH)-one (47 mg, 0.11 mmol) and 2-bromo-5-methylpyridine (94 mg, 0.55 mmol) were stirred dry toluene (3 mL)/DMSO (1 mL) and degassed with a nitrogen stream as the temperature was increased to 1000C. Palladium tetrakistriphenylphosphine (13 mg, 0.011 mmol) was added and the reaction was maintained at 1000C under a nitrogen atmosphere for 16 h. Upon cooling, the mixture was purified by Combiflash chromatography (12 g ISCO column eluting with methylene chloride and methanol/ammonia (10:1); 100% methylene chloride to 20% methanol/ammonia over 30 min at 40 mL/min). Concentration of the appropriate fractions provided the free base. Conversion to the hydrochloride salt as in Example 1 gave the title compound (48 mg, 51%) as a white solid: mp 230-2340C; 1H NMR (500 MHz, CD3OD) δ 8.56 (s, IH), 8.23 (s, IH), 7.91-7.89 (m, 2H), 7.81-7.76 (m, 3H), 7.54-7.51 (dd, J = 8.9, 2.0 Hz, IH), 7.24 (s, IH) 7.17-7.15 (dd, J = 7.5, 1.9 Hz, IH), 4.92- 4.84 (br m, 2 H), 3.74-3.55 (br m, 2H), 3.25-3.06 (br m, 4 H), 2.43 (s, 3H), 2.09-1.88 (br m, 4H); ESI MS m/z 400 [M + H]+; HPLC (Method C) >99% (AUC), tR = 10.3 min.
Example 31
Preparation of 4-(2-Chloro-4-(trifluoromethv0phenv0- 1 -( 1 -(2-(pyrrolidin- 1 -vDethvO- IH- indazol-5-yl)pyridin-2(lH)-one hydrochloride
Figure imgf000069_0001
[00114] Following the procedure of Example 30, but substituting 2-chloro-5- (trifluoromethyl)iodobenzene for 2-bromo-5-methylpyridine, the title compound (21 mg, 38%) was prepared as a white solid: 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.94-7.91 (m, 2H), 7.83 (d, J = 8.9 Hz, IH), 7.80-7.77 (m, 2H), 7.70 (d, J = 8.0 Hz, IH), 7.57-7.55 (dd, J= 8.9, 1.9 Hz, IH), 6.72 (d, J= 1.5 Hz, IH), 6.63-6.62 (dd, J= 7.0, 1.8 Hz, IH), 4.89 (t, J= 5.6, 2H), 3.87 (t, J= 5.6, 2H), 3.80-3.57 (br m, 2 H), 3.20-3.02 (br m, 2H), 2.23-194 (br m, 4H); ESI MS m/z 487 [M + H]+; HPLC (Method C) 98.4% (AUC), tR = 13.9 min.
Example 32
Preparation of 1 -( 1 -(2-(Pyrrolidin- 1 -yDethyl)- lH-indazo 1-5 -yl)-4-(quino lin-2-yl)pyridin- 2(l/-/)-one trihydrochloride
Figure imgf000069_0002
[00115] Following the procedure of Example 30, but substituting 2-chloroquinoline for 2- bromo-5-methylpyridine, the title compound (12 mg, 21%) was prepared as a yellow solid: mp 220-2250C; 1H NMR (500 MHz, CD3OD) δ 8.97 (d, J = 8.6 Hz, IH), 8.33 (d, J = 8.6 Hz, IH), 8.30 (d, J= 8.1 Hz, IH), 8.28 (s, IH), 8.25 (d, J= 8.1 Hz, IH), 8.10-8.07 (m, IH), 7.98-7.96 (m, 2H), 7.91 (d, J= 8.0 Hz, IH), 7.87 (d, J= 7.0 Hz, IH), 7.60-7.58 (dd, J= 8.9, 2.0 Hz, IH), 7.40 (d, J = 1.9 Hz, IH), 7.25-7.23 (dd, J = 7.1, 2.0 Hz, IH), 4.91 (t, J = 5.9, 2H), 3.88 (t, J= 5.9, 2H), 3.75-3.70 (m, 2 H), 3.21-3.15 (m, 2H), 2.19-2.16 (m, 2H), 2.05-
2.01 (m, 2H); ESI MS m/z 436 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.1 min.
Example 33
Preparation of 4-(5-Chloropyridin-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2( lHVone trihydrochloride
Figure imgf000070_0001
[00116] Following the procedure of Example 30, but substituting 2-bromo-5- chloropyridine for 2-bromo-5-methylpyridine, the title compound (14 mg, 26%) was prepared as a yellow solid: 1H NMR (500 MHz, CD3OD) δ 8.77-8.69 (m, IH), 8.28 (d, J = 1.7 Hz, IH), 8.05 (d, J = 8.5 Hz, IH), 8.03-8.01 (dd, J = 8.5, 2.4 Hz, IH), 7.93 (d, J = 1.4 Hz, IH), 7.84 (d, J = 8.9 Hz, IH), 7.81 (d, J = 7.1 Hz, IH), 7.57-7.55 (dd, J = 8.9, 1.9 Hz, IH), 7.33 (d, J = 1.6 Hz, IH), 7.24-7.22 (dd, J = 7.2, 2.0 Hz, IH), 4.90 (t, J = 5.8, 2 H), 3.89 (t, J = 5.8, 2H), 3.75-3.71 (m, 2 H), 3.22-3.12 (m, 2H), 2.21-2.16 (m, 2H), 2.05-2.02 (m, 2H); ESI MS m/z 420 [M + H]+; HPLC (Method C) >99% (AUC), tR = 11.6 min.
Example 34
Preparation of 1 -(I -(2-(Pyrrolidin- 1 -yDethyO- lH-indazol-5-yl)-4-(5- (trifluoromethyl)pyridin-2-yl)pyridin-2( lHVone trihydrochloride
Figure imgf000071_0001
[00117] Following the procedure of Example 30, but substituting 2-bromo-5- trifluoromethylpyridine for 2-bromo-5-methylpyridine, the title compound (10 mg, 16%) was prepared as a yellow solid: 1H NMR (500 MHz, CD3OD) δ 9.05 (s, IH), 8.29-8.27 (dd, J = 8.4, 2.2 Hz, IH), 8.27 (s, IH), 8.22 (d, J = 8.4 Hz, IH), 7.93 (d, J = 1.6 Hz, IH), 7.84- 7.82 (2 overlapping d, J= 8.9, 7.0 Hz, 2H) 7.57-7.55 (dd, J= 8.9, 1.9 Hz, IH), 7.40 (d, J = 1.8 Hz, IH), 7.27-7.25 (dd, J= 7.1, 1.9 Hz, IH), 4.89 (t, J= 5.8, 2H), 3.87 (t, J= 5.8, 2H), 3.74-3.70 (m, 2H), 3.21-3.15 (m, 2H), 2.19-2.15 (m, 2H), 2.04-2.00 (m, 2H); ESI MS m/z 454 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.4 min.
Example 35
Preparation of 1 -( 1 -(2-(Pyrrolidin- 1 -yDethyl)- lH-indazo 1-5 -yl)-4-(quinazo lin-2-yl)pyridin- 2(l/-/)-one dihydrochloride
Figure imgf000071_0002
[00118] Following the procedure of Example 30, but substituting 2-chloroquinazoline for 2-bromo-5-methylpyridine, the title compound (12 mg, 23%) was prepared as a green solid: 1H NMR (500 MHz, CD3OD) δ 9.66 (s, IH), 8.27 (s, IH), 8.19-8.15 (m, 2H), 8.09-8.06 (m, IH), 7.95 (d, J= 1.6 Hz, IH), 7.91 (d, J= 1.6 Hz, IH) 7.86-7.81 (m, 3H), 7.69-7.67 (dd, J = 7.1, 1.8 Hz, IH), 7.60-7.57 (dd, J = 8.9, 2.0 Hz, IH), 4.90 (t, J = 5.8, 2H), 3.88 (t, J = 5.9, 2H), 3.75-3.70 (m, 2 H), 3.21-3.16 (br m, 2H), 2.20-2.17 (m, 2H), 2.04-2.00 (m, 2H); ESI MS m/z 437 [M + H]+; HPLC (Method C) >99% (AUC), tR = 11.6 min.
Example 36
Preparation of 1 -(I -(2-(Pyrrolidin- 1 -vOethvO- lH-indazol-5-yl)-4-(6- (trifluoromethyl)pyridazin-3-yl)pyridin-2(l/-/)-one hydrochloride
Figure imgf000072_0001
[00119] Following the procedure of Example 30, but substituting 3-chloro-6- (trifluoromethyl)pyridazine for 2-bromo-5-methylpyridine, the title compound (14 mg, 26%) was prepared as a yellow solid: 1H NMR (500 MHz, CD3OD) δ 8.53 (d, J = 8.9 Hz, IH), 8.28-8.26 (m, 2H), 7.95 (d, J = 1.6 Hz, IH), 7.91 (d, J = 7.0 Hz, IH), 7.86 (d, J = 8.9 Hz, IH), 7.58-7.56 (dd, J = 8.9, 1.9 Hz, IH) 7.44 (d, J = 1.6 Hz, IH), 7.36-7.34 (dd, J = 7.1, 2.0 Hz, IH), 4.89 (t, J= 5.9, 2H), 3.88 (t, J= 5.9, 2H), 3.72-3.70 (br m, 2 H), 3.21-3.15 (br m, 2H), 2.19-2.17 (m, 2H), 2.09-2.07 (m, 2H); ESI MS m/z 455 [M + H]+; HPLC (Method C) 97.7% (AUC), to = 11.6 min.
Example 37
Preparation of l-(l-(2-Morpholinoethyl)-l/-f-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
a) 5-Iodo-lH-indazole
Chemical Formula: C7H5IN2 Exact Mass: 243.95
Figure imgf000072_0002
Molecular Weight: 244.03 [00120] A solution of 4-iodo-2-methylaniline (10.0 g, 42.9 mmol) in glacial acetic acid (400 niL) was treated with a solution Of NaNO2 (2.96 g, 42.9 mmol) in water (10 mL). After stirring for 6 hours, the mixture was concentrated to dryness and dissolved in ethyl acetate (EtOAc). Filtration through a pad of silica gel (EtOAc) provided the title compound (10.4 g, 99%) as a deep purple solid: ESI MS m/z 245 [M+ H]+.
b) 1 -(2,2-Dimethoxyethyl)-5-iodo- lH-indazole
Chemical Formula: CnHi3IN2O2 Exact Mass: 332 Molecular Weight: 332.14
Figure imgf000073_0001
[00121] To a solution of 5-iodio-l/f-indazole (8.28 g, 33.9 mmol) in DMSO (104 mL) was added 2-bromoacetaldehyde dimethyl acetal (7.9 mL, 68 mmol) and CS2CO3 (44.1 g, 136 mmol). The reaction was stirred at 4O0C for 18 h; then the reaction was diluted with H2O (100 mL) and EtOAc (175mL). The partitioned material was extracted with EtOAc (4 x 175 mL). The organics were washed with brine (2 x 100 mL), dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (silica gel, hexanes with 0.1% Et3N /EtOAc with 0.1% Et3N, 100:0 to 90:10) gave the title compound (4.49 g, 46%) as a light orange powder: 1H NMR (500 MHz, CDCl3) δ 8.07 (d, J= 1.0 Hz, IH), 7.92 (d, J= 0.5 Hz, IH), 7.60 (dd, J = 9.0, 1.5 Hz, IH), 7.28 (d, J = 9.0 Hz, IH), 4.71 (t, J = 5.5 Hz, IH), 4.44 (d, J= 5.5 Hz, 2H), 3.33 (s, 6H).
c) 4-(4-(Trifluoromethyl)phenyl)pyridine 1 -oxide
Chemical Formula: C12H8F3NO
Exact Mass: 239.06
Figure imgf000073_0002
Molecular Weight: 239.19
[00122] A solution of 4-(trifluoromethyl)phenylboronic acid (1.78 g, 9.37 mmol) in DME (10 mL) and aqueous K2CO3 (12 mL, 1.8 M) was degassed with argon for 20 minutes. Triphenylphosphine (797 mg, 3.04 mmol), palladium(II) acetate (174 mg, 0.775 mmol), and 4-chloropyridine 1-oxide (1.00 g, 7.72 mmol) were added sequentially, and the mixture was heated to reflux under an argon atmosphere. After stirring at reflux for 14 h, the mixture was allowed to cool and then filtered. The filtrate was diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 rnL). The organics were dried over Na2SO4, filtered and concentrated to dryness. Purification by flash column chromatography (silica gel, CH2Cl2/Me0H, 95:5) gave the title compound (465 mg, 25%) as white crystals: 1U NMR (500 MHz, CDCl5) δ 8.29 (d, J= 7.2 Hz, 2H), 7.75 (d, J= 8.3 Hz, 2H), 7.69 (d, J= 8.2 Hz, 2H), 7.52 (d, J= 7.2 Hz, 2H); ESI MS m/z 240 [M + H]+.
d) 4-(4-(Trifluoromethyl)phenyl)pyridin-2( lH)-one
Chemical Formula: 012HgF3NO
// \ // X NH Exact Mass: 239.06
Molecular Weight: 239.19
[00123] A stirred solution of 4-(4-(trifluoromethyl)phenyl)pyridine 1 -oxide (465 mg, 1.94 mmol) in Ac2O (10 mL) under nitrogen atmosphere was heated from 110 to 1300C over 4.5 h. Then the mixture was heated at reflux for 2 h and then allowed to cool. The mixture was concentrated to dryness and then treated with MeOH and water (10 mL, 1 :1). After stirring at room temperature for 6 h, the mixture was heated to reflux for 2.5 h. The mixture was allowed to cool and was concentrated to dryness. Purification by flash column chromatography (silica gel, CH2Cl2/Me0H, 95:5) gave the title compound (336 mg, 72%) as a light brown powder: 1H NMR (500 MHz, CDCl5) δ 12.67 (br s, IH), 7.76 (d, J= 8.2 Hz, 2H), 7.69 (d, J= 8.3 Hz, 2H), 7.47 (d, J= 6.8 Hz, IH), 6.80 (d, J= 1.6 Hz, IH), 6.54 (dd, J = 6.8, 1.8 Hz, IH); ESI MS m/z 240 [M + H]+.
e) l-(l-(2,2-Dimethoxyethyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one
Chemical Formula: C23H20F3N3O3
Exact Mass: 443.15 Molecular Weight: 443.42
Figure imgf000074_0001
[00124] A suspension of l-(2,2-dimethoxyethyl)-5-iodo-lH-indazole (458 mg, 1.38 mmol), 4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one (330 mg, 1.38 mmol), Cs2CO3 (997 mg, 3.06 mmol), 8-hydroxyquinoline (42 mg, 0.29 mmol) and CuI (311 g, 163 mmol) in DMSO (5 niL) was evacuated for 30 minutes under high vacuum then backfilled with argon. The mixture was stirred under argon at 1150C for 15 h and then allowed to cool. The mixture was diluted with 10% NH4OH in H2O (40 mL) and extracted with EtOAc (4 x 50 mL). The organics were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated to dryness. Purification by flash chromatography (silica gel, CH2Cl2ZMeOH, 95:5) followed by a second purification by flash chromatography (silica gel, CH2Cl2/Me0H, 97.5:2.5) gave the title compound (482 mg, 79%) as a light brown solid: 1H NMR (500 MHz, CDCl5) δ 8.08 (s, IH), 7.75-7.73 (m, 5H), 7.61 (d, J = 8.9 Hz, IH), 7.52 (d, J = 7.1 Hz, IH), 7.45 (dd, J = 8.9, 1.8 Hz, IH), 6.92 (d, J= 1.8 Hz, IH), 6.52 (dd, J = 7.1, 1.9 Hz, IH), 4.78 (t, J = 5.3 Hz, IH), 4.52 (d, J = 5.3 Hz, 2H), 3.39 (s, 6H); ESI MS m/z 444 [M + H]+.
f) 2-(5-(2-Oxo-4-(4-(trifluoromethyl)phenyl)pyridin- 1 (2H)-yl)- lH-indazol- 1 - yl)acetaldehyde
Figure imgf000075_0001
[00125] A solution of l-(l-(2,2-dimethoxyethyl)-lH-indazol-5-yl)-4-(4- (trifluoromethyl)- phenyl)pyridin-2(l/-/)-one (480 mg, 1.08 mmol) in THF (10 mL) was treated with aqueous HCl (9 mL, 2.0 M). The solution was heated to reflux for 1 h, allowed to cool and then treated with H2O (100 mL). The resulting solids were isolated by filtration, washed with H2O and dried under high vacuum for 16 h to give the title compound (1.00 g, 64%) as a light brown solid: ESI MS m/z 398 [M + H]+.
g) l-(l-(2-Morpholinoethyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one Chemical Formula: C25H23F3N4O2
Exact Mass: 468.18 Molecular Weight: 468.47
Figure imgf000076_0001
[00126] To a solution of 2-(5-(2-oxo-4-(4-(trifluoromethyl)phenyl)pyridin-l(2H)-yl)-l/f- indazol-l-yl)acetaldehyde (84 mg, 0.21 mmol) in CH2Cl2 (4.0 mL), MeOH (1.0 niL) and AcOH (0.50 mL) was added morpholine (0.06 mL, 0.7 mmol) and picoline-borane complex (25 mg, 0.23 mmol). After stirring at ambient temperature under nitrogen atmosphere for 1.5 h, the solution was treated with 1 N HCl (10.0 mL) and stirred vigorously for 30 minutes. The mixture was made basic with saturated aqueous NaHCO3 (25 mL) and extracted with CH2Cl2 (3 x 25 mL). The organics were dried over Na2SO4, filtered and concentrated to dryness. Purification by flash chromatography (silica gel, CH2Cl2/Me0H, 97:3) gave the title compound (33 mg, 33%) as an off-white solid: 1H NMR (500 MHz, CDCl5) δ 8.14 (d, J= 0.7 Hz, IH), 7.95 (d, J= 8.1 Hz, 2H), 7.85-7.77 (m, 5H), 7.48 (dd, J = 8.9, 1.9 Hz, IH), 6.95 (d, J= 1.8 Hz, IH), 6.85 (dd, J= 7.1, 2.0 Hz, IH), 4.63 (t, J= 6.5 Hz, 2H), 3.64-3.62 (m, 4H), 2.91 (t, J= 6.5 Hz, 2H), 2.54-2.52 (m, 4H); ESI MS m/z 469 [M + H]+.
h) l-(l-(2-Morpholinoethyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one hydrochloride
Chemical Formula: C25H23F3N4O2
Exact Mass: 468.18 Molecular Weight: 468.47
Figure imgf000076_0002
[00127] A solution of l-(l-(2-morpholinoethyl)-lH-indazol-5-yl)-4-(4-(trifluoro-methyl)- phenyl)pyridin-2(lH)-one (32 mg, 0.068 mmol) in CH2Cl2 (2 niL) was treated with anhydrous HCl in diethyl ether (0.07 mL, 0.07 mmol, 1.0 M). After stirring at ambient temperature for 15 min, the reaction mixture was diluted with Et2O (20 mL). The resulting solids were collected by filtration and dried in a vacuum oven to yield the title compound (24 mg, 69%) as an off-white powder: mp 248-2500C (decomposition); 1H NMR (500 MHz, DMSO-J6) δ 10.58 (br s, IH), 8.28 (s, IH), 8.02 (d, J = 8.0 Hz, 2H), 7.92-7.84 (m, 5H), 7.55 (d, J= 9.0 Hz, IH), 6.90 (d, J = 2.0 Hz, IH), 6.75 (dd, J = 7.0, 2.0 Hz, IH), 4.95 (br, 2H), 4.02-3.99 (m, 2H), 3.72-3.70 (m, 4H), 3.56-3.54 (m, 2H), 3.20 (br, 2H); ESI MS m/z 469 [M+ H]+; HPLC (Method B) 98.9% (AUC), tR = 14.8 min.
Example 38
Preparation of (S)- 1 -( 1 -(2-(3 -hydroxypyrrolidin- 1 -vOethvO- lH-indazo 1-5 - yP)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
Chemical Formula: C25H23F3N4O2
Exact Mass: 468.18 Molecular Weight: 468.47
Figure imgf000077_0001
[00128] Following the procedure of Example 37, but substituting (5)-pyrrolidin-3-ol for morpholine, the title compound (10 mg, 10%) was prepared as an off-white powder: 1H NMR (500 MHz, DMSO-J6) δ 10.65 (br s, 0.4H), 10.41 (br s, 0.6H), 8.28 (br s, IH), 8.02 (d, J= 8.0 Hz, 2H), 7.94-7.85 (m, 5H), 7.54 (d, J= 9.0 Hz, IH), 6.90 (d, J= 2.0 Hz, IH), 6.75 (dd, J= 7.5, 2.0 Hz, IH), 5.49 (br s, IH), 4.90-4.88 (m, 2H), 4.43-4.37 (m, IH), 3.78-3.71 (m, 2H), 3.60 (br, IH), 3.41-3.34 (m, IH), 3.17-3.13 (m, IH), 2.99-2.97 (m, IH), 2.25-2.22 (m, IH), 1.95-1.81 (m, IH); ESI MS m/z 469 [M + H]+; HPLC (Method B) 98.2% (AUC), tR = 15.7 min; Optical Rotation [α]23 5 D -4.3° (c 1.00, Methanol). Example 39
Preparation of (R)- 1 -( 1 -(2-(3 -hydroxypyrro lidin- 1 - vOethvO- lH-indazo 1-5 - yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lH)-one hydrochloride
HO
Chemical Formula: C25H23F3N4O2
Exact Mass: 468.18 Molecular Weight: 468.47
Figure imgf000078_0001
[00129] Following the procedure of Example 37, but substituting (i?)-pyrrolidin-3-ol for morpholine, the title compound (23 mg, 22%) was prepared as an off-white powder: 1H NMR (500 MHz, DMSO-J6) δ 10.40 (br s, 0.3H), 10.31 (br s, 0.5H), 8.28 (d, J = 6.5 Hz, IH), 8.02 (d, J= 8.0 Hz, 2H), 7.93-7.84 (m, 5H), 7.54 (d, J= 8.0 Hz, IH), 6.90 (s, IH), 6.75 (d, J = 7.0 Hz, IH), 5.50 (br s, IH), 4.90-4.88 (m, 2H), 4.44-4.38 (m, IH), 3.79-3.72 (m, 2H), 3.61-3.60 (m, IH), 3.42-3.40 (m, IH), 3.14-3.13 (m, IH), 3.01-2.99 (m, IH), 2.25- 2.24 (m, IH), 1.95-1.81 (m, IH); ESI MS m/z 469 [M + H]+; HPLC (Method B) 96.7% (AUC), tR = 13.9 min; Optical Rotation [α]23 5 D +3.7° (c 1.00, Methanol).
Example 40
Preparation of (R)- 1 -( 1 -(2-(2-(Hvdroxymethv0pyrrolidin- 1 - vOethvO- lH-indazo 1-5 - yP)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
Chemical Formula: C26H26ClF3N4O2"
Exact Mass: 518.17 Molecular Weight: 518.96
Figure imgf000078_0002
[00130] Following the procedure of Example 37, but substituting (i?)-pyrrolidin-2- ylmethanol for morpholine, the title compound (88 mg, 58%) was prepared as a yellow powder: 1U NMR (500 MHz, DMSO-J6) δ 9.85 (s, IH), 8.28 (s, IH), 8.04-8.01 (m, 2H), 7.93-7.85 (m, 5H), 7.54 (dd, J= 8.8, 2.0 Hz, IH), 6.90 (d, J= 1.9 Hz, IH), 6.75 (dd, J= 7.2, 2.0 Hz, IH), 4.95-4.85 (m, 2H), 3.97-3.93 (m, IH), 3.81-3.76 (m, IH), 3.71-3.63 (m, 3H), 3.58-3.52 (m, IH), 3.16-3.10 (m, IH), 2.14-2.05 (m, IH), 2.04-1.97 (m, IH), 1.89-1.81 (m, IH), 1.78-1.70 (m, IH); ESI MS m/z 483 [M + H]+; HPLC (Method B) 96.4% (AUC), tR = 15.0 min.
Example 41
Preparation of (S)- 1 -(I -(2-(2-(Hvdroxymethyl)pyrrolidin- 1 -vDethvO- l/f-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
Chemical Formula: C26H26ClF3N4O2"
Exact Mass: 518.17 Molecular Weight: 518.96
Figure imgf000079_0001
[00131] Following the procedure of Example 37, but substituting (5)-pyrrolidin-2- ylmethanol for morpholine, the title compound (76 mg, 50%) was prepared as a yellow powder: 1H NMR (500 MHz, DMSO-J6) δ 9.88 (s, IH), 8.28 (s, IH), 8.04-8.00 (m, 2H), 7.92-7.83 (m, 5H), 7.54 (dd, J= 9.0, 1.9 Hz, IH), 6.90 (d, J= 2.0 Hz, IH), 6.75 (dd, J= 7.2, 2.0 Hz, IH), 4.95-4.85 (m, 2H), 3.98-3.91 (m, IH), 3.82-3.75 (m, IH), 3.70-3.63 (m, 3H), 3.58-3.52 (m, IH), 3.15-3.09 (m, IH), 2.12-2.05 (m, IH), 2.03-1.97 (m, IH), 1.92-1.82 (m, IH), 1.78-1.71 (m, IH); ESI MS m/z 483 [M + H]+; HPLC (Method B) 96.4% (AUC), tR = 16.0 min.
Example 42
Preparation of 1 -( 1 -(2-Dimethylamino)ethyl)- lH-indazo 1-5 -yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride mical Formula: C23H22ClF3N4O Exact Mass: 462.14
Molecular Weight: 462.90
Figure imgf000080_0001
[00132] Following the procedure of Example 37, but substituting dimethylamine for morpholine, the title compound (45 mg, 100%) was prepared as a white powder: mp 222- 224°C; 1H NMR (500 MHz, DMSO-J6) δ 9.76 (br s, IH), 8.28 (s, IH), 8.02-8.01 (m, 2H), 7.92-7.84 (m, 5H), 7.55-7.53 (m, IH), 6.89 (d, J= 2.0 Hz, IH), 6.76-6.74 (m, IH), 4.89 (t, J = 6.0 Hz, 2H), 3.65-3.64 (m, 2H), 2.86 (s, 6H); ESI MS m/z All [M + H]+; HPLC (Method B) >99% (AUC), tR = 14.8 min.
Example 43
Preparation of 1 -(I -(2-(Piperazin- 1 -vOethvO- l/f-indazol-5-vD-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride a) 1 -(3-Methyl-4-nitrophenyl)-4-(4-(trifluoromethyl)phenyl)pyridin-2( lH)-one
Chemical Formula: C19H13F3N2O3
Exact Mass: 374.09 Molecular Weight: 374.31
Figure imgf000080_0002
[00133] To a solution of 5-fluoro-2-nitrotoluene (0.27 g, 2.25 mmol) in DMF (4.0 mL) was added 4-(4-(trifluoromethyl)phenyl-pyridin-2(lH)-one (0.45 g, 1.87 mmol) and Cs2CO3 (0.67 g, 2.1 mmol), and the reaction was heated to 85 0C for 18 h. The reaction mixture was cooled, H2O (20 mL) was added, and the mixture was stirred for 20 min. The resulting solids were collected by filtration and washed with H2O (1OmL). Flash chromatography (silica gel, hexanes/EtOAc, 10:1 to 3:1) yielded the title compound (0.45 g, 65%) as a yellow solid: 1H NMR (300 MHz, DMSO-J6) δ 8.33 (d, J= 5.4 Hz, IH), 8.12-7.89 (m, 5H), 7.65-7.60 (m, 2H), 7.33-7.22 (m, 2H), 2.55 (s, 3H).
b) l-(4-Amino-3-methylphenyl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one Chemical Formula: C19H15F3N2O
Exact Mass: 344.11 Molecular Weight: 344.33
Figure imgf000081_0001
[00134] A solution of l-(3-methyl-4-nitrophenyl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one (0.46 g, 1.2 mmol) in 9:1 EtOH/H2O (20 niL) was treated with iron powder (0.62 g, 11.1 mmol) and NH4Cl (33 mg, 0.61 mmol), and the resulting suspension was heated at reflux for 18 h. The reaction mixture was filtered, while hot, through Celite® with portions OfCH2Cl2 (2 x 25mL) and MeOH (2 x 25mL). The filtrates were combined and made basic with 1 N NaOH (1OmL). The solution was extracted with CH2Cl2 (2 x 4OmL) and the combined organic extracts were dried over Na2SO4, filtered and concentrated to provide the title compound (0.409 g, 100%) as a yellow solid: 1H NMR (500 MHz, DMSO-J6) δ 7.97 (d, J= 8.0 Hz, 2H), 7.85 (d, J= 8.5 Hz, 2H), 7.68 (d, J= 7.0 Hz, IH), 6.96 (s, IH), 6.92 (dd, J = 8.5, 2.0 Hz, IH), 6.80 (d, J =1.5 Hz, IH), 6.67 (d, J = 8.0 Hz, IH), 6.63 (dd, J = 7.0, 2.0 Hz, IH), 5.11 (s, 2H), 2.09 (s, 3H).
c) l-(l/-f-Indazol-5-yl) -4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one
Chemical Formula: C19H12F3N3O
Exact Mass: 355.09 Molecular Weight: 355.31
Figure imgf000081_0002
[00135] A solution of l-(4-amino-3-methylphenyl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one (0.405 g, 1.22 mmol) in AcOH (12.5 mL) was treated with a solution OfNaNO2 (84.0 mg, 1.22 mmol) in H2O (0.4 mL) and stirred at ambient temperature for 18 h. The reaction mixture was concentrated. Flash chromatography (silica gel,
CH2Cl2/MeOH/NH4OH, 90:9:1) yielded the title compound (0.330 g, 76%) as a yellow solid: 1H NMR (500 MHz, DMSO-J6) δ 13.28 (s, IH), 8.17 (s, IH), 8.01 (d, J= 8.0 Hz, 2H), 7.88-7.85 (m, 4H), 7.64 (d, J= 8.8 Hz, IH), 7.39 (dd, J= 8.8, 2.0 Hz, IH), 6.89 (d, J= 2.0 Hz, IH), 6.72 (dd, J= 7.0, 2.0 Hz, IH). d) 1 -( 1 -(2-Chloroethyl)- lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2( lH)-one
Chemical Formula: C21H15ClF3N3O Exact Mass: 417.09
Molecular Weight: 417.81
Figure imgf000082_0001
[00136] To a solution of l-(l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one (120 mg, 0.338 mmol) in DMSO (2.0 mL) was added 2-bromo-l-chloroethane (485 mg, 3.38 mmol) and CS2CO3 (330 mg, 1.01 mmol), and the reaction was stirred at ambient temperature for 3 h. The reaction was diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 mL). The extracts were washed with brine (25 mL), dried over Na2SO4, and concentrated. Flash chromatography (Biotage 25+M column, CH2Cl2/Me0H, 99:1 to 98:2) yielded the title compound (72 mg, 51%): 1H NMR (500 MHz, CDCl3) δ 8.11 (s, IH), 7.76- 7.73 (m, 5H), 7.59-7.47 (m, 3H), 6.92-6.91 (m, IH), 6.53-6.51 (m, IH), 4.74-4.71 (m, 2H), 4.00-3.98 (m, 2H).
e) 1 -( 1 -(2-(Piperazin- 1 -yl)ethyl)- lH-indazo 1-5 -yl)-4-(4-(trifluoromethyl)phenyl)pyridin-
hemical Formula: C25H24F3N5O Exact Mass: 467.19
Molecular Weight: 467.49
Figure imgf000082_0002
[00137] To a solution of l-(l-(2-chloroethyl)-l/f-indazol-5-yl)-4-(4-
(trifluoromethyl)phenyl)pyridin-2(lH)-one (72 mg, 0.17 mmol) in DMF (2.0 mL) was added K2CO3 (0.12 g, 0.86 mmol), piperazine (0.29 g, 3.4 mmol) and KI (29 mg, 0.17 mmol), and the reaction was heated to 50 0C for 3 h. The reaction was cooled, diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 mL). The combined organic extracts were washed with brine (25 mL), dried over Na2SO4, filtered and concentrated. Flash chromatography (silica gel, CH2Cl2/MeOH/NH4OH, 30:1 :0 to 10:1 :0.2) provided the title compound (65 mg, 80%): 1H NMR (500 MHz, CDCl3) δ 8.05 (s, IH), 7.75-7.74 (m, 5H), 7.57-7.76 (m, 3H), 6.92 (d, J= 2.0 Hz, IH), 6.52 (d, J= 7.0 Hz, IH), 4.55 (t, J= 7.0 Hz, 2H), 2.89-2.87 (m, 6H), 2.51- 2.50 (m, 4H).
f) 1 -( 1 -(2-(Piperazin- 1 -yl)ethyl)- l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-
2(lH)-one hydrochloride
Chemical Formula: C25H25ClF3N5O Cl Exact Mass: 503.17 Molecular Weight: 503.95
Figure imgf000083_0001
[00138] According to the procedure of Example 37 step h, except substituting l-(l-(2- (piperazin-l-yl)ethyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one for l-(l-(2-morpholinoethyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)-phenyl)pyridin-2(l/-/)- one, the title compound (32 mg, 83%) was prepared as a white solid: mp 222-224 0C (decomposition); 1H NMR (500 MHz, DMSO-J6) δ 8.24 (s, IH), 8.02-8.01 (m, 2H), 7.92- 7.85 (m, 5H), 7.51 (d, J = 8.6 Hz, IH), 6.89 (d, J = 2.0 Hz, IH), 6.75 (dd, J = 7.1, 1.8 Hz, IH), 4.87-4.85 (m, 2H), 3.71-3.60 (m, 6H), 3.38-3.31 (m, 4H); ESI MS m/z 468 [M + H]+; HPLC (Method B) 96.2% (AUC), tR = 13.9 min.
Example 44
Preparation of l-(l-(3-(Dimethylamino)propyl)-l/-f-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
a) 1 -( 1 -(3 -(Dimethylamino)propyl)- 1 H-indazo 1-5 -yl)-4-(4-(trifluoromethyl)phenyl) pyridin-2( lH)-one CH,
Chemical Formula: C24H23F3N4O
Exact Mass: 440.18 Molecular Weight: 440.46
Figure imgf000084_0001
[00139] To a solution of l-(l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one (278 mg, 0.783 mmol) in DMSO (4.0 niL) was added 3-bromo-l-chloropropane (1.23 g, 7.83 mmol) and CS2CO3 (765 mg, 2.35 mmol), and the reaction was stirred at ambient temperature for 18 h. The reaction was diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 mL). The extracts were washed with brine (25 mL), dried over Na2SO4, and concentrated. Flash chromatography (silica gel, CH2Cl2ZMeOH, 100:1) yielded l-(l-(3- chloropropyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one. To a solution of 1 -( 1 -(3 -chloropropyl)- lH-indazo 1-5 -yl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one (84 mg, 0.19 mmol) in DMF (1.0 mL) was added K2CO3 (0.13 g, 0.97 mmol), dimethylamine (1.94 mL, 3.88 mmol, 2.0 M in THF) and KI (32 mg, 0.19 mmol), and the reaction was heated to 45 0C for 18 h. The reaction mixture was cooled, diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 mL). The combined organic extracts were washed with brine (25 mL), dried over Na2SO4, filtered and concentrated. Flash chromatography (Biotage 25+ M column, CH2Cl2/MeOH/NH4OH, 50:1 :0 to 20:1 :0.1) provided the title compound (54 mg, 63%): 1H NMR (500 MHz, CDCl3) δ 8.06 (s, IH), 7.75-7.70 (m, 5H), 7.60-7.43 (m, 3H), 6.92-6.91 (m, IH), 6.53-6.51 (m, IH), 4.49 (t, J = 6.8 Hz, 2H), 2.29 (t, J= 6.8 Hz, 2H), 2.23 (s, 6H), 2.13-2.08 (m, 2H).
b) l-(l-(3-(Dimethylamino)propyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl) pyridine-2(l/-/)-one hydrochloride Chemical Formula: C24H24ClF3N4O •HC1 Exact Mass: 476.16 Molecular Weight: 476.92
Figure imgf000085_0001
[00140] According to the procedure of Example 37 step h, except substituting l-(l-(3- (dimethylamino)propyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one for l-(l-(2-morpholinoethyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)-phenyl)pyridin-2(lH)- one, the title compound (48 mg, 79%) was prepared as a white solid: mp 209-211 0C; 1H NMR (500 MHz, DMSO-J6) δ 9.85 (br s, IH), 8.21 (s, IH), 8.01 (d, J= 8.0 Hz, 2H), 7.88- 7.84 (m, 5H), 7.50-7.48 (m, IH), 6.89-6.88 (m, IH), 6.74 (dd, J= 7.0, 1.7 Hz, IH), 4.57 (t, J= 6.5 Hz, 2H), 3.10 (t, J= 7.5 Hz, 2H), 2.75 (s, 6H), 2.26-2.22 (m, 2H); ESI MS m/z 441 [M + H]+; HPLC (Method B) 98.9% (AUC), tR = 15.4 min.
Example 45
Preparation of 1 -(I -(2-(Piperazin- 1 -vOethvO- lH-indazol-5-vD-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
a) 1 -( 1 -(3 -(Pyrrolidin- 1 -yl)propyl)- lH-indazol-5 -yl)-4-(4(trifluoromethyl)phenyl) pyridin-
Chemical Formula: C26H25F3N4O
Exact Mass: 466.20 Molecular Weight: 466.50
Figure imgf000085_0002
[00141] To a solution of l-(lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one (278 mg, 0.783 mmol) in DMSO (4.0 mL) was added 3-bromo-l-chloropropane (1.23 g,
7.83 mmol) and CS2CO3 (765 mg, 2.35 mmol), and the reaction was stirred at ambient temperature for 18 h. The reaction mixture was diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 mL). The extracts were washed with brine (25 mL), dried over Na2SO4, and concentrated. Flash chromatography (silica gel, CH2Cl2ZMeOH, 100:1) yielded the intermediate l-(l-(3-chloropropyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one. To a solution of l-(l-(3-chloropropyl)-lH-indazol-5-yl)-4-(4-
(trifluoromethyl)phenyl)pyridin-2(lH)-one (78 mg, 0.18 mmol) in DMF (1.0 mL) was added K2CO3 (0.12 g, 0.90 mmol), pyrrolidine (130 mg, 1.8 mmol) and KI (30 mg, 0.18 mmol), and the reaction was heated to 50 0C for 18 h. The reaction mixture was cooled and diluted with H2O (25 mL). The resulting solids were collected by filtration and washed with H2O (10 mL). The collected solid was dissolved in EtOAc (2OmL) and washed with brine (1OmL). The washed organic solution was dried over Na2SO4, filtered and concentrated to provide the title compound (77 mg, 91%): 1H NMR (500 MHz, CDCl3) δ 8.05 (s, IH), 7.77- 7.74 (m, 5H), 7.60-7.42 (m, 3H), 6.92 (d, J = 2.0 Hz, IH), 6.53-6.51 (m, IH), 4.51 (t, J = 7.0 Hz, 2H), 2.48-2.44 (m, 6H), 2.16-2.12 (m, 2H), 1.79-1.77 (m, 4H).
b) 1 -(I -(3-(Pyrrolidin- 1 -yl)propyl)- l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl) pyridine- 2(lH)-one hydrochloride
Chemical Formula: C26H26ClF3N4O •HC1 Exact Mass: 502.17 Molecular Weight: 502.96
Figure imgf000086_0001
[00142] According to the procedure of Example 37 step h, except substituting l-(l-(3- (pyrrolidin-l-yl)propyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one for l-(l-(2-morpholinoethyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)-phenyl)pyridin-2(l/-/)- one, the title compound (54 mg, 67%) was prepared as a yellow solid: 1H NMR (500 MHz, DMSO-J6) δ 10.26 (br s, IH), 8.21 (s, IH), 8.01 (d, J = 8.1 Hz, 2H), 7.88-7.84 (m, 5H), 7.49 (dd, J= 8.8, 1.9 Hz, IH), 6.89 (d, J= 1.9 Hz, IH), 6.74 (dd, J= 7.1, 2.0 Hz, IH), 4.59 (t, J= 7.0 Hz, 2H), 3.54-3.51 (m, 2H), 3.19-3.15 (m, 2H), 2.99-2.93 (m, 2H), 2.30-2.24 (m, 2H), 2.01-1.95 (m, 2H), 1.89-1.84 (m, 2H); ESI MS m/z 467 [M + H]+; HPLC (Method B) 96.8% (AUC), tR = 15.9 min.
Example 46
Preparation of l-(l-(3-(Amino)propyl)-lH-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
a) l-(l-(3-Aminopropyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one
Chemical Formula: C22H19F3N4O
Exact Mass: 412.15 Molecular Weight: 412.41
Figure imgf000087_0001
[00143] To a solution of l-(l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)- one (278 mg, 0.783 mmol) in DMSO (4.0 rnL) was added 3-bromo-l-chloropropane (1.23 g, 7.83 mmol) and CS2CO3 (765 mg, 2.35 mmol), and the reaction was stirred at ambient temperature for 18 h. The reaction was diluted with H2O (25 mL) and extracted with EtOAc (3 x 25 mL). The extracts were washed with brine (25 mL), dried over Na2SO4, and concentrated. Flash chromatography (silica gel, CH2Cl2ZMeOH, 100:1) yielded l-(l-(3- chloropropyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one. To a solution of 1 -( 1 -(3 -chloropropyl)- lH-indazo 1-5 -yl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one (84 g, 0.20 mmol) in DMF (1.5 mL) was added potassium phthalimide (41 mg, 0.22 mmol) and the reaction was heated to 100 0C for 4 h. The reaction was allowed to cool, diluted with H2O (20 mL) and extracted with CH2Cl2 (3 x 25 mL). The combined organic extracts were washed with brine (25 mL), dried over Na2SO4, and concentrated. Flash chromatography (Biotage 25 + M column, CH2Cl2/Me0H, 50:1 to 40:1) yielded 2-(3-(5-(2- oxo-4-(4-(trifluoromethyl)phenyl)pyridin- 1 (2H)-yl)- lH-indazol- 1 -yl)propyl)isoindoline- 1,3- dione. 2-(3-(5-(2-oxo-4-(4-(trifluoromethyl)phenyl)pyridin- 1 (2H)-yl)- 1/f-indazol- 1 - yl)propyl)isoindoline-l,3-dione (73 mg, 0.13 mmol) was dissolved in ethanolamine (1.0 mL) and stirred at ambient temperature for 18 h. The reaction was concentrated. Flash column chromatography (Biotage 25+ M column, CH2Cl2/MeOH/NH4OH, 30:1 :0.1 to 10:1 :0.2) yielded l-(l-(3-aminopropyl)-lH-indazol-5-yl)-4-(4-
(trifluoromethyl)phenyl)pyridin-2(lH)-one. l-(l-(3-aminopropyl)-lH-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2(lH)-one was dissolved in CH2Cl2 (1.0 mL) and Et3N (30 μL, 0.22 mmol), and di-tert-buty\ dicarbonate (26 mg, 0.12mmol) were added. The resulting solution was stirred at ambient temperature for 18 h. The reaction was concentrated. Flash column chromatography (silica gel, CH2Cl2/Me0H, 40:1) afforded tert- butyl-3-(5-(2-oxo-4-(4-(trifluoromethyl)phenyl)pyridin- 1 (2H)-yl)- 1/f-indazol- 1 - yl)propylcarbamate. J'ert-butyl-3-(5-(2-oxo-4-(4-(trifluoromethyl)phenyl)pyridin- 1 (2H)-yl)- l/-f-indazol-l-yl)propylcarbamate was dissolved in CH2Cl2 (1.0 mL) and TFA (25 μL, 0.33 mmol). The resulting solution was stirred at ambient temperature for 18 h. The reaction was diluted with CH2Cl2 (1OmL) and washed with saturated NaHCCh. The organic solution was dried over Na2SO4, filtered and concentrated. Flash column chromatography (silica gel, CH2Cl2/MeOH/NH4OH, 30:1 :0.1 to 20:1 :0.1) yielded the title compound (16 mg, 29%): 1H NMR (500 MHz, CDCl3) δ 8.03 (s, IH), 7.77-7.74 (m, 5H), 7.58-7.44 (m, 3H), 6.92 (d, J = 1.5 Hz, IH), 6.53 (dd, J= 7.5, 2.0 Hz, IH), 4.54 (t, J= 7.0 Hz, 2H), 2.70 (t, J= 7.0 Hz, 2H), 2.10-2.04 (m, 2H).
b) l-(l-(3-Aminopropyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one hydrochloride
Chemical Formula: C22H20ClF3N4O
Exact Mass: 448.13 Molecular Weight: 448.87
Figure imgf000088_0001
[00144] According to the procedure of Example 37 step h, except substituting l-(l-(3- aminopropyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one for l-(l-(2- morpholinoethyl)-l/-f-indazol-5-yl)-4-(4-(trifluoromethyl)-phenyl)pyridin-2(l/-/)-one, the title compound (8.4 mg, 52%) was prepared as a yellow powder: mp 222-224 0C
(decomposition); 1H NMR (500 MHz, DMSO-J6) δ 8.21 (s, IH), 8.01 (d, J = 8.5 Hz, 2H), 7.89-7.80 (m, 8H), 7.50-7.48 (m, IH), 6.89 (d, J = 1.5 Hz, IH), 6.74 (dd, J = 7.0, 2.0 Hz, IH), 4.58 (t, J= 6.5 Hz, 2H), 2.84-2.80 (m, 2H), 2.16-2.12 (m, 2H); ESI MS m/z 413 [M + H]+; HPLC (Method B) 97.0% (AUC), tR = 14.9 min.
Example 47
Preparation of (S)- 1 -( 1 -(Pyrro lidin-2- ylmethvQ- lH-indazo 1-5 - yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
Chemical Formula: C24H22ClF3N4O Exact Mass: 474.14 Molecular Weight: 474.91
Figure imgf000089_0001
[00145] A mixture of l-(lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one (200 mg, 0.56 mmol), (S)-tert-buty{ 2-(bromomethyl)pyrrolidine-l-carboxylate (GRA-B- 188) (298 mg, 1.12 mmol), and cesium carbonate (732 mg, 2.25 mmol) in methyl sulfoxide (5 mL) was stirred overnight at ambient temperature. The reaction mixture was diluted with water (20 mL) and extracted with dichloromethane (2 x 30 mL). The combined organic extracts were washed with brine (20 mL), dried (Na2SO4), and concentrated under reduced pressure. Isolation of the desired regioisomer was performed by column chromatography (silica gel, Et2O to 9:1 Et2O/90:9:l Et2O /MeOH /coned NH4OH). To the desired regioisomer (100 mg, 0.18 mmol) was added dichloromethane (10 mL) and trifluoroacetic acid (2 mL). After stirring at ambient temperature for 2 h, the reaction was brought to pH = 9 with IN NaOH. The organic layer was washed with brine (10 mL), dried (Na2SO4), and concentrated under reduced pressure. The dried residue (87 mg, 0.20 mmol) was dissolved in dichloromethane (10 mL) and HCl (1.25 M solution in methanol, 0.22 mL, 0.17 mmol) was added. The mixture was concentrated under reduced pressure and then triturated with dichloromethane/hexanes and the obtained solid was triturated with diethyl ether. The solid was dried to afford the title compound (43 mg, 16%) as an off-white solid: mp 241-245 0C; 1H NMR (500 MHz, DMSO-J6) δ 9.05 (br s, 2H), 8.28 (s, IH), 8.03-7.85 (m, 7H), 7.51 (d, J = 8.5 Hz, IH), 6.89 (s, IH), 6.75 (d. J = 7.0 Hz, IH), 4.80 (d, J = 6.0 Hz, 2H), 3.99-3.95 (m,lH), 3.30-3.23 (m, IH), 3.16-3.10 (m, IH), 2.08-2.03 (m, IH), 1.99-1.96 (m, IH), 1.91-1.85 (m, IH), 1.74-1.70 (m, IH); ESI MS m/z 439 [M + H]+; HPLC (Method B)
98.7% (AUC), tR = 15.5 min; [αf 23 D +20.5° (c 0.13, Methanol).
Example 48
Preparation of (i?)-l-(l-(3-(Dimethylamino)-2-hydroxypropyl)-lH-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
a) (S)- 1 -( 1 -(Oxiran-2-ylmethyl)- lH-indazo 1-5 -yl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one
Chemical Formula: C22H16F3N3O2
Exact Mass: 41 1.12 Molecular Weight: 41 1.38
Figure imgf000090_0001
[00146] A mixture of l-(l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one (750 mg, 2.11 mmol), (i?)-(-)-glycidyl nosylate (657 mg, 2.53 mmol) and cesium carbonate (1.03 g, 3.17 mmol) in methyl sulfoxide (6 mL) was stirred at ambient temperature overnight. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were washed with brine (20 mL), dried (Na2SO4), and concentrated under reduced pressure. Purification by column chromatography (silica gel, ethyl acetate) gave the title compound (480 mg, 55%) as a yellow solid: 1H NMR (300 MHz, CDCl3) δ 8.09 (d, J = 0.9 Hz, IH), 7.76-7.72 (m, 5H), 7.65 (d, J= 9.0 Hz, IH), 7.52 (d, J= 7.2 Hz, IH), 7.47 (dd, J= 9.0, 2.1 Hz, IH), 6.92 (d, J = 1.8 Hz, IH), 6.53 (dd, J= 6.9, 1.8 Hz, IH), 4.79 (dd, J= 15, 3.0 Hz, IH), 4.48 (dd, J= 15.3, 5.7 hz, IH), 3.41-3.37 (m, IH), 2.88 (app t, J= 4.5 Hz, IH), 2.60 (dd, J= 4.5, 2.4 Hz, IH).
b) (i?)-l-(l-(3-(Dimethylamino)-2-hydroxypropyl)-lH-indazol-5-yl)-4-(4-
(trifluoromethyl)phenyl)pyridin-2( lH)-one hydrochloride Chemical Formula: C24H24ClF3N4O2
Exact Mass: 492.15 Molecular Weight: 492.92
Figure imgf000091_0001
[00147] To (5)-l-(l-(oxiran-2-ylmethyl)-l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)- pyridin-2(lH)-one (200 mg, 0.48 mmol) in tetrahydrofuran (5 mL) was added LiClO4 (775 mg, 7.3 mmol) followed by dimethylamine (2.4 ml of a 2 M solution in tetrahydrofuran, 4.9 mmol). The reaction mixture was heated in a 45 0C oil bath for 4.5 h. The reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml), washed with water (25 mL) and brine (25 mL), dried (Na2SO4), and concentrated under reduced pressure. The material was purified by column chromatography (silica gel, 9:1 CH2Cl2ZMeOH to 90:9:1 CH2Cl2/Me0H /cone. NH4OH). The dried residue (160 mg, 0.35 mmol) was dissolved in dichloromethane (10 mL) and HCl (1.25 M solution in methanol, 0.31 mL, 0.43 mmol) was added. The mixture was concentrated under reduced pressure and dried to afford the title compound (190 mg, 80%) as a yellow solid: mp 210-213 0C; 1H NMR (500 MHz, DMSO-J6) δ 9.50 (br s, IH), 8.22 (s, IH), 8.02-8.00 (m, 2H), 7.89-7.84 (m, 5H), 7.48 (dd, J = 9.0, 2.0 Hz, IH), 6.89 (d, J = 2.0 Hz, IH), 6.74 (dd, J = 7.0 Hz, 2.0 Hz, IH), 6.01 (br s, IH), 4.58-4.49 (m, 2H), 4.40 (br s, IH), 3.28-3.24 (m, IH), 3.15-3.10 (m, IH), 2.81 (d, J = 4.5 Hz, 3H), 2.78 (d, J= 4.5 Hz, 3H); ESI MS m/z 457 [M + H]+; HPLC (Method B) 96.5% (AUC), tR = 14.4 min; [α]23 D +10.4° (c 0.11, Methanol).
Example 49
Preparation of (i?)-l-(l-(2-Hydroxy-3-(pyrrolidin-l-yl)propyl)-lH-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride Chemical Formula: C26H26ClF3N4O2
Exact Mass: 518.17 Molecular Weight: 518.96
Figure imgf000092_0001
[00148] To (5)-l-(l-(oxiran-2-ylmethyl)-l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)- pyridin-2(lH)-one (200 mg, 0.48 mmol) in tetrahydrofuran (5 niL) was added LiClO4 (775 mg, 7.28 mmol) followed by pyrrolidine (0.41 mL, 4.9 mmol). The reaction mixture was heated in a 45 0C oil bath for 4.5 h. The reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml), washed with water (25 mL) and brine (25 mL), dried (Na2SO4), and concentrated under reduced pressure. The material was purified by column chromatography (silica gel, 9:1 CH2Cl2/Me0H to 90:9:1 CH2Cl2/Me0H /cone. NH4OH). The dried residue (197 mg, 0.41 mmol) was dissolved in dichloromethane (10 mL) and HCl (1.25 M solution in methanol, 0.36 mL, 0.45 mmol) was added. The mixture was concentrated under reduced pressure and dried to afford the title compound (194 mg, 78%) as a yellow solid: mp 116-119 0C; 1H NMR (500 MHz, DMSO-J6) δ 9.65 (br s, IH), 8.21 (s, IH), 8.02-8.00 (m, 2H), 7.89-7.82 (m, 5H), 7.47 (dd, J = 9.0, 2.0 Hz, IH), 6.89 (d, J= 2.0 Hz, IH), 6.74 (dd, J= 7.0, 2.0 Hz, IH), 5.75 (br s, IH), 4.57-4.47 (m, 2H), 4.33 (br s, IH), 3.56-2.90 (br m, 6H), 1.89 (br s, 4H); ESI MS m/z 483 [M + H]+; HPLC (Method B) >99% (AUC), tR = 14.5 min; [α]23 D +10.8° (c 0.11, Methanol).
Example 50
Preparation of (S)- 1 -(I -(3-(Dimethylamino)-2-hydroxypropyl)- l/-f-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
a) (R)- 1 -( 1 -(Oxiran-2-ylmethyl)- lH-indazol-5 -yl)-4-(4-(trifluoromethyl)phenyl)pyridin- 2(lH)-one Chemical Formula: C22H16F3N3O2
Exact Mass: 41 1.12 Molecular Weight: 41 1.38
Figure imgf000093_0001
[00149] A mixture of l-(l/f-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one (1.1 g, 3.1 mmol), (5)-(+)-glycidyl nosylate (0.96 g, 3.7 mmol) and cesium carbonate (1.5 g, 4.6 mmol) in methyl sulfoxide (10 mL) was stirred at ambient temperature overnight. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (2 x 50 mL). The combined organic extracts were washed with brine (20 mL), dried (Na2SO4), and concentrated under reduced pressure. Purification by column chromatography (silica gel, ethyl acetate) gave the title compound (510 mg, 40%) as a yellow solid: 1H NMR (500 MHz, CDCl3) δ 8.08 (s, IH), 7.76-7.73 (m, 5H), 7.64 (d, J = 9.0 Hz, IH), 7.51 (d, J = 7.0 Hz, IH), 7.47 (dd, J = 9.0, 2.0 Hz, IH), 6.92 (d, J= 2.0 Hz, IH), 6.52 (dd, J = 7.5, 2.0 Hz, IH), 4.78 (dd, J = 15.5, 3.5 Hz, IH), 4.48 (dd, J = 15.0, 5.5 Hz, IH), 3.41-3.38 (m, IH), 2.88 (app t, J= 4.5 Hz, IH), 2.60 (dd, J= 5.0, 3.0 Hz, IH).
b) (5)-l-(l-(3-(Dimethylamino)-2-hydroxypropyl)-lH-indazol-5-yl)-4-(4-
(trifluoromethyl)phenyl)pyridin-2( lH)-one hydrochloride
Chemical Formula: C24H24ClF3N4O2
Exact Mass: 492.15 Molecular Weight: 492.92
Figure imgf000093_0002
[00150] To (i?)-l-(l-(oxiran-2-ylmethyl)-lH-indazol-5-yl)-4-(4-(trifiuoromethyl)- phenyl)pyridin-2(l/-/)-one (230 mg, 0.56 mmol) in tetrahydrofuran (5 mL) was added LiClO4 (892 mg, 8.38 mmol) followed by dimethylamine (2.8 ml of a 2 M solution in tetrahydrofuran, 5.6 mmol). The reaction mixture was heated in a 45 0C oil bath for 3.75 h. The reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml), washed with water (25 mL) and brine (25 mL), dried (Na2SO4), and concentrated under reduced pressure. The material was purified by column chromatography (silica gel, 9:1 CH2Cl2/Me0H to 90:9:1 CH2Cl2/Me0H /cone. NH4OH). The dried residue (198 mg, 0.43 mmol) was dissolved in dichloromethane (2 mL), and HCl (1.25 M solution in methanol, 0.38 mL, 0.47 mmol) was added. The mixture was concentrated under reduced pressure and dried to afford the title compound (198 mg, 71%) as a yellow solid: 1H NMR (500 MHz, DMSO-J6) δ 9.42 (br s, IH), 8.20 (s, IH), 8.02-8.00 (m, 2H), 7.88-7.80 (m, 5H), 7.46 (dd, J = 8.5, 1.5 Hz, IH), 6.89 (d, J = 2.0 Hz, IH), 6.74 (dd, J= 7.0, 2.0 Hz, IH), 5.60 (br s, IH), 4.56-4.52 (m, IH), 4.48-4.44 (m, IH), 4.27 (br s, IH), 2.92-2.88 (br m, 2H), 2.58 (br s, 6H); ESI MS m/z 457 [M + H]+; HPLC (Method B) >99% (AUC), tR = 14.5 min; [α]23 D - 9.5° (c 0.14, Methanol).
Example 51
Preparation of (S)- 1 -(I -(2-Hydroxy-3-(pyrrolidin- 1 -yPpropyl)- l/-f-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lHVone hydrochloride
Chemical Formula: C26H26ClF3N4O2
Exact Mass: 518.17 Molecular Weight: 518.96
Figure imgf000094_0001
[00151] To (i?)-l-(l-(oxiran-2-ylmethyl)-lH-indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)- pyridin-2(l/-/)-one (250 mg, 0.61 mmol) in tetrahydrofuran (5 mL) was added LiClO4 (969 mg, 9.11 mmol) followed by pyrrolidine (0.51 mL, 6.1 mmol). The reaction mixture was heated in a 45 0C oil bath for 3.75 h. The reaction mixture was cooled to ambient temperature, diluted with dichloromethane (50 ml) and washed with water (25 mL), brine (25 mL), dried (Na2SO4), and concentrated under reduced pressure. The material was purified by column chromatography (silica gel, 9:1 CH2Cl2MeOH to 90:9:1 CH2Cl2MeOH /cone. NH4OH). The dried residue (233 mg, 0.48 mmol) was dissolved in dichloromethane
(2 mL), and HCl (1.25 M solution in methanol, 0.42 mL, 0.53 mmol) was added. The mixture was concentrated under reduced pressure and dried to afford the title compound (240 mg, 76%) as a yellow solid: 1H NMR (500 MHz, DMSO-J6) δ 9.60 (br s, IH), 8.20 (s, IH), 8.02-8.00 (m, 2H), 7.89-7.80 (m, 5H), 7.46 (dd, J = 8.5, 1.5 Hz, IH), 6.89 (d, J = 2.0 Hz, IH), 6.74 (dd, J = 7.5, 2.0 Hz, IH), 4.57-4.53 (m, IH), 4.49-4.46 (m, IH), 4.27 (br s, IH), 3.25-2.75 (br m, 6H), 1.85 (br s, 4H); ESI MS m/z 483 [M + H]+; HPLC (Method B) >99% (AUC), tR = 15.1 min; [α]23 D -11.3° (c 0.11, Methanol).
Example 52
Preparation of (+)- 1 -(I -(2-((l£4iS)-2-Oxa-5-azabicvclor2.2.1 lheptan-5-yl)ethyl)- IH- indazol-5-yl)-4-(4-(trifluoromethyl)phenyl)pyridin-2(lH)-one hydrochloride
a) 5-Bromo- 1 -(2-chloroethyl)- lH-indazole Chemical Formula: C9H8BrClN2 Exact Mass: 257.96 Molecular Weight: 259.53
Figure imgf000095_0001
[00152] l-Bromo-2-chloroethane (2.9 mL, 35 mmol) was added to a suspension of 5- bromo-lH-indazole (4.302 g, 21.95 mmol) and Cs2CO3 (28.56 g, 87.81 mmol) in DMSO (50 mL) under N2. The resulting suspension was stirred at 25 0C for 6 h. H2O (50 mL) was added, and the resulting suspension was cooled in an ice bath. The suspension was filtered, and the solid was dried under reduced pressure to afford a pink powder. Flash chromatography on silica gel (19:1 to 5:2 hexanes/EtOAc) gave the title compound (3.094 g, 54%) as a white solid: 1H NMR (500 MHz, CDCl3) δ 8.00 (br s, IH), 7.90 (d, J = 1.5 Hz, IH), 7.50 (dd, J= 9.0, 1.5 Hz, IH), 7.38 (d, J= 9.0 Hz, IH), 4.68 (t, J= 6.3 Hz, 2H), 3.98 (t, J= 6.3 Hz, 2H).
b) 5-(2-(5-Bromo- lH-indazol- 1 -yl)ethyl)-(15',45)-2-oxa-5-azabicyclo[2.2.1 ]heptane Chemical Formula: C14H16BrN3O Exact Mass: 321.05 Molecular Weight: 322.20
Figure imgf000095_0002
[00153] 5-Bromo- 1 -(2-chloroethyl)- lH-indazo Ie (183 mg, 0.703 mmol) was added to a suspension of (15*,45)-(+)-5-aza-2-oxabicyclo[2.2.1]heptane hydrochloride (286 mg, 2.11 mmol), K2CO3 (485 mg, 3.52 mmol) and KI (117 mg, 0.703 mmol) in DMF (10 mL) under N2. The resulting suspension was stirred at 90 0C for 19 h. The suspension was cooled, and H2O (10 mL) was added. The aqueous solution was extracted with EtOAc, and the combined organic extracts were washed with brine. The organic solution was dried over Na2SO4 and concentrated under reduced pressure to afford clear viscous oil. Flash chromatography on silica gel (100:0 to 0:100 hexanes/(9:0.9:0.1 CH2Cl2/MeOH/NH4OH)) gave the title compound (88 mg, 39%) as a clear oil: 1U NMR (300 MHz, CDCl3) δ 7.95- 7.92 (m, IH), 7.87-7.84 (m, IH), 7.45 (dd, J = 8.7, 1.8 Hz, IH), 7.34 (d, J = 8.7 Hz, IH), 4.43 (t, J= 6.7 Hz, 2H), 4.34 (br s, IH), 3.92 (d, J= 7.8 Hz, IH), 3.56 (dd, J= 7.8, 1.5 Hz, IH), 3.34 (br s, IH), 3.15-3.01 (m, 2H), 2.92 (d, J= 21.9 Hz, IH), 2.81 (dd, J= 9.9, 1.5 Hz, IH), 2.47 (br d, J= 9.9 Hz, IH), 1.79-1.60 (m, IH).
c) (+)-l-(l-(2-((llS,45)-2-Oxa-5-azabicyclo[2.2.1]heptan-5-yl)ethyl)-lH-indazol-5-yl)-4-(4- (trifluoromethyl)phenyl)pyridin-2( lH)-one hydrochloride
Chemical Formula: C26H24ClF3N4O2
Exact Mass: 516.15 Molecular Weight: 516.94
Figure imgf000096_0001
[00154] A suspension of 4-(4-(trifluoromethyl)phenyl)pyridin-2(l/-/)-one (54 mg, 0.23 mmol), 5-(2-(5-bromo-lH-indazol-l-yl)ethyl)-(15',45)-2-oxa-5-azabicyclo[2.2.1]heptane (88 mg, 0.27 mmol), CuI (52 mg, 0.27 mmol), 8-hydroxyquinoline (7.0 mg, 0.045 mmol) and Cs2SO4 (81 mg, 0.25 mmol) in DMSO (5 mL) was degassed under reduced pressure for 45 min. The suspension was put under Ar and heated at 130 0C with stirring for 22 h. The suspension was cooled, NH4OH was added and the resulting suspension was stirred at 25 0C for 30 min. The suspension was passed through a plug of silica gel with 4:1 CH2C12/(9: 1 MeOH/NH4OH). The phases of the filtrate were separated and the aqueous phase was extracted with CH2Cl2. The combined organic extracts were washed with brine, dried over Na2SO4 and concentrated under reduced pressure to afford a viscous oil. Flash chromatography on silica gel (hexanes/(9:0.9:0.1 CH2Cl2/MeOH/NH4OH) 100:0 to 9:1) gave 13 mg of a viscous oil. 1.0 M HCl in Et2O (0.03 mL, 0.03 mmol) was added to a solution of the viscous oil in CH2Cl2 (10 niL) under N2, and the mixture was stirred at 25 0C for 1 h. The solution was concentrated to afford the title compound (14 mg, 12%) as an off- white powder: mp 148-150 0C; 1H NMR (500 MHz, DMSO-J6) δ 10.62-10.50 (m, 0.4H), 10.21-10.09 (m, 0.6H), 8.28 (s, IH), 8.02 (d, J= 8.0 Hz, 2H), 7.96-7.80 (m, 5H), 7.55 (d, J = 9.0 Hz, IH), 6.90 (s, IH), 6.75 (dd, J = 7.0, 1.5 Hz, IH), 4.93-4.86 (m, 2H), 4.70 (s, 0.6H), 4.61-4.52 (m, 1.4H), 4.22 (d, J= 11.0 Hz, 0.4H), 4.07-4.01 (m, 0.6H), 3.92-3.79 (m, 1.2H), 3.77 (d, J = 8.5 Hz, 0.6H), 3.71-3.60 (m, 0.8H), 3.59-3.54 (m, 0.6H), 3.51-3.43 (m, 0.4H), 3.22 (d, J= 12.0 Hz, 0.6 H), 2.64 (s, 0.4H), 2.39-2.36 (m, IH), 2.12 (d, J= 11.5 Hz, 0.4H), 2.05-1.99 (m, IH); ESI MS m/z 481 [M + H]+; [α]24 D + 15.0 °(c 1.00, MeOH).
Example 53
Preparation of 4-(4-Chloro-2-fluorophenyD- 1 -(I -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol-5- yl)pyridin-2(l/-/)-one hydrochloride
Chemical Formula: C24H23Cl2FN4O
Exact Mass: 472.12 Molecular Weight: 473.37
Figure imgf000097_0001
[00155] Following the procedure of Example 1, but substituting 2-fluoro-4- chlorophenylboronic acid for phenylboronic acid, the title compound (26.7 mg, 28%) was prepared as orange solid: mp 215-230 0C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.92 (d, J = 1.8 Hz, IH), 7.83 (d, J = 8.9 Hz, IH), 7.77 (d, J = 8.9 Hz, IH), 7.65 (t, J = 8.2 Hz, IH), 7.56-7.53 (dd, J= 8.9, 1.9 Hz, IH), 7.65 (m, 2H), 6.85 (m, IH), 6.76-6.73 (dd, J= 7.1, 1.8 Hz, IH), 4.89 (t, J = 5.7 Hz, 2H), 3.88 (t, J = 5.7 Hz, 2H), 3.74-3.70 (m, 2H), 3.20- 3.15 (m, 2H), 2.19-2.16 (m, 2H), 2.04-2.01 (m, 2H); ESI MS m/z 437 [M + H]+; HPLC (Method C) >99% (AUC), tR = 13.1 min.
Example 54
Preparation of 4-(4-(Trifluoromethyl)-2-fluorophenyl)- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- IH- indazol-5-yl)pyridin-2(lH)-one hydrochloride Chemical Formula: C25H23ClF4N4O
Exact Mass: 506.15 Molecular Weight: 506.92
Figure imgf000098_0001
[00156] Following the procedure of Example 1, but substituting 2-fluoro-4- methoxyphenylboronic acid for phenylboronic acid, the title compound (40.8 mg, 40%) was prepared as a brown solid: mp 80-95 0C; 1H NMR (500 MHz, CD3OD) δ 8.27 (d, J = 0.5 Hz, IH), 7.93 (d, J= 1.7 Hz, IH), 7.86-7.79 (m, 3H), 7.68-7.65 (m, 2H), 7.57-7.54 (dd, J = 8.9, 1.9 Hz, IH), 6.89 (s, IH), 6.77-6.75 (m, IH), 4.89 (t, J= 5.7 Hz, 2H), 3.88 (t, J= 5.7 Hz, 2H), 3.73-3.69 (m, 2H), 3.21-3.15 (m, 2H), 2.20-2.13 (m, 2H), 2.06-2.00 (m, 2H); ESI MS m/z 471 [M + H]+; HPLC (Method C) 96.1% (AUC), tR = 13.6 min.
Example 55
Preparation of 4-(4-Methoxy-2-fluorophenyD- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(l/-/)-one hydrochloride
Chemical Formula: C25H26ClFN4O2
Exact Mass: 468.17 Molecular Weight: 468.95
Figure imgf000098_0002
[00157] Following the procedure of Example 1, but substituting 2-fluoro-4- methoxyphenylboronic acid for phenylboronic acid, the title compound (82.4 mg, 86%) was prepared as a brown-orange solid: mp 230-240 0C; 1H NMR (500 MHz, CD3OD) δ 8.26 (d, J= 0.7 Hz, IH), 7.91 (d, J= 1.6 Hz, IH), 7.82 (d, J= 8.9 Hz, IH), 7.72 (d, J= 7.2 Hz, IH), 7.58 (t, J= 8.9 Hz, IH), 7.55-7.53 (dd, J= 8.9, 1.9 Hz, IH), 6.93-6.91 (dd, J= 8.7, 2.4 Hz, IH), 6.88-6.86 (dd, J= 13.2, 2.4 Hz, IH), 6.83 (s, IH), 6.77-6.75 (m, IH), 4.89 (t, J= 5.7 Hz, 2H), 3.88 (m, 5H), 3.73-3.69 (m, 2H), 3.20-3.14 (m, 2H), 2.19-2.13 (m, 2H), 2.05-2.01
(m, 2H); ESI MS m/z 433 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.6 min. Example 56
Preparation of 4-(4-Methoxy-2-chlorophenyD- 1 -(I -(2-(pyrroridin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Chemical Formula: C25H26Cl2N4O2
Exact Mass: 484.14 Molecular Weight: 485.41
Figure imgf000099_0001
[00158] Following the procedure in Example 1, but substituting 2-chloro-4- methoxyphenylboronic acid for phenylboronic acid, the title compound (67.3 mg, 37%) was prepared as a brown-orange solid: mp 225-235 0C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.92 (d, J = 1.8 Hz, IH), 7.82 (d, J = 8.9 Hz, IH), 7.71 (d, J= 7.0 Hz, IH), 7.56-7.54 (dd, J= 8.9, 1.9 Hz, IH), 7.42 (d, J= 8.6, Hz, IH), 7.14 (d, J= 2.5 Hz, IH), 7.04-7.02 (dd, J= 8.6, 2.5 Hz, IH), 6.68 (d, J= 1.8 Hz, IH), 6.65-6.63 (dd, J= 7.0, 1.9 Hz, IH), 4.89 (t, J = 5.7 Hz, 2H), 3.88-3.87 (m, 5H), 3.72-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.21-2.13 (m, 2H), 2.06-2.01 (m, 2H); ESI MS m/z 449 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.9 min.
Example 57
Preparation of 4-(4-EthoxyphenyD- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazo 1-5 -yPpyridin- 2(l/-/)-one hydrochloride
Chemical Formula: C26H29ClN4O2
Exact Mass: 464.20 Molecular Weight: 464.99
Figure imgf000099_0002
[00159] Following the procedure in Example 1, but substituting 4-ethoxyphenylboronic acid for phenylboronic acid, the title compound (54.5 mg, 23%) was prepared as a orange solid: mp 255-265 0C; 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.90 (d, J = 1.9 Hz, IH), 7.82 (d, J= 8.9 Hz, IH), 7.74-7.72 (m, 3H), 7.55-7.52 (dd, J= 8.9, 1.9 Hz, IH), 7.07- 7.04 (m, 2H), 6.90-6.88 (m, 2H), 4.89 (t, J= 5.7 Hz, 2H), 4.12 (q, J= 7.0 Hz, 2H), 3.88 (t, J = 5.7 Hz, 2H), 3.73-3.69 (m, 2H), 3.20-3.15 (m, 2H), 2.19-2.14 (m, 2H), 2.04-2.00 (m, 2H), 1.43 (t, J = 7.0 Hz, 3H); ESI MS m/z 429 [M + H]+; HPLC (Method C) >99% (AUC), IR = 13.1 min.
Example 58
Preparation of 4-(4-(Trifluoromethoxy)-2-fluorophenyl)- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- IH- indazol-5-yl)pyridin-2(lH)-one hydrochloride
Chemical Formula: C25H23ClF4N4O2
Exact Mass: 522.14 Molecular Weight: 522.92
Figure imgf000100_0001
[00160] Following the procedure in Example 30, but substituting l-bromo-2-fluoro-4- (trifluoromethoxy)benzene for 2-bromo-5-methylpyridine, the title compound (24.1 mg, 19%) was prepared as a yellow solid: mp 215-225 0C; 1H NMR (500 MHz, CD3OD) δ 8.26 (d, J = 0.4 Hz, IH), 7.92 (d, J = 1.8 Hz, IH), 7.82 (d, J = 8.9 Hz, IH), 7.78-7.75 (m, 2H), 7.56-7.54 (dd, J = 8.9, 1.9 Hz, IH), 7.33-7.30 (m, 2H), 6.86 (s, IH), 6.75-6.73 (m, IH), 4.89 (t, J = 5.8, 2H), 3.87 (t, J = 5.8, 2H), 3.71 (m, 2H), 3.19 (m, 2H), 2.16-2.04 (m, 4H); ESI MS m/z 487 [M + H]+; HPLC (Method C) >99% (AUC), tR = 14.3 min.
Example 59
Preparation of 4-(4-(Trifluoromethoxy)-2-methylphenyl)- 1 -( 1 -(2-(pyrrolidin- 1 -yDethyl)- IH- indazol-5-yl)pyridin-2(lH)-one hydrochloride Chemical Formula: C26H26ClF3N4O2
Exact Mass: 518.17 Molecular Weight: 518.96
Figure imgf000101_0001
[00161] Following the procedure in Example 30, but substituting l-bromo-2-fluoro-4- (trifluoromethoxy)benzene for 2-bromo-5-methylpyridine, the title compound (49.0 mg, 21%) was prepared as a orange solid: mp 185-195 0C; 1H NMR (500 MHz, CD3OD) δ 8.27 (s, IH), 7.93 (d, J = 1.7 Hz, IH), 7.83 (d, J = 8.9 Hz, IH), 7.75 (d, J = 7.0 Hz, IH), 7.57- 7.55 (dd, J = 8.9, 1.9 Hz, IH), 7.41 (d, J = 8.4 Hz, IH), 7.27 (s, IH), 7.23 (d, J = 8.4 Hz, IH), 6.61 (d, J = 1.7 Hz, IH), 6.56-6.54 (dd, J = 7.0, 1.8 Hz, IH), 4.90 (t, J = 5.7, 2H), 3.88 (t, J= 5.7, 2H), 3.74-3.69 (m, 2H), 3.21-3.15 (m, 2H), 2.43 (s, 3H), 2.19-2.16 (m, 2H) 2.04-2.00 (m, 2H); ESI MS m/z 483 [M + H]+; HPLC (Method C) >99% (AUC), tR = 14.7 min.
Example 60
Preparation of 4-( 1 -Methyl- lH-indazo 1-5 -yl)- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyl)- lH-indazo 1-5 - vDpyridine 2(\H)-one hydrochloride:
a) 5-Bromo-l -methyl- IH- indazo Ie
Beilstein Registry Number 127881
Chemical Formula: C8H7BrN2
Exact Mass: 209.98 Molecular Weight: 211.06
Figure imgf000101_0002
[00162] To a solution of 5-bromo-lH-indazole (1.0 g, 5.07 mmol) in DMSO (15 mL) was added iodomethane (0.41 mL, 6.6 mmol) and K2CO3 (3.4 g, 25 mmol). The reaction was stirred at room temperature for 3 h; then the reaction was diluted with methylene chloride (100 mL) and washed with a 5 % LiCl solution (4x). The organics were washed with brine (50 mL), dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (40 g ISCO column, hexanes/EtOAc, 100:0 to 70:30) gave the title compound (532 mg, 50%) as a white powder: 1U NMR (500 MHz, CDCl3) δ 7.91 (s, IH), 7.87 (d, J = 1.7 Hz, IH), 7.47-7.44 (dd, J= 8.9, 1.7 Hz, IH), 7.28 (d, J= 8.8 Hz, IH), 4.06 (s, 3H).
b) 4-(l -Methyl- lH-indazol-5-yl)- 1 -(I -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)pyridine 2(lH)-one hydrochloride
Chemical Formula: C26H27ClN6O
Exact Mass: 474.19 Molecular Weight: 474.99
Figure imgf000102_0001
[00163] Following the procedure in Example 30, but substituting 5-bromo-l -methyl- IH- indazole for 2-bromo-5-methylpyridine, the title compound (18.0 mg, 11%) was prepared as a orange solid: mp 55-65 0C; 1H NMR (500 MHz, CD3OD) δ 8.27 (d, J= 0.9 Hz, IH), 8.23 (d, J = 0.9 Hz, IH), 8.14 (d, J = 0.9 Hz, IH), 7.93 (d, J = 1.4 Hz, IH), 7.84 (t, J = 8.9 Hz, 2H), 7.80-7.78 (dd, J = 5.8, 1.9 Hz, IH), 7.72 (d, J = 8.9 Hz, IH), 7.58-7.55 (dd, J = 8.9, 1.9 Hz, IH), 7.01-6.99 (m, 2H), 4.90 (t, J = 5.7, 2H), 4.12 (s, 3H), 3.88 (t, J = 5.7, 2H), 3.74-3.70 (m, 2H), 3.21-3.16 (m, 2H), 2.21-2.14 (m, 2H) 2.07-1.99 (m, 2H); ESI MS m/z 439 [M + H]+; HPLC (Method C) 96.5% (AUC), tR = 11.0 min.
Example 61
1 -(I -(3-(Pyrrolidin- 1 -yPpropyl)- lH-indazol-5-yl)-4-(5-(trifluoromethyl)pyridin-2- yl)pyridin-2(lH)-one hydrochloride
a) 2'-Methoxy-5-(trifluoromethyl)-2,4'-bipyridine
Chemical Formula: C12H9F3N2O
Exact Mass : 254.07 Molecular Weight: 254.21
Figure imgf000102_0002
[00164] 2-Bromo-5-trifluoromethylpyridine (410 mg, 2.13 mmol), 2-methoxy-4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine (500 mg, 1.81 mmol), K2CO3 (749 mg, 5.43 mmol) and [l,r-Bis-(diphosphenylphosphino)ferrocene] dichloropalladium(II) (PdC^dppf) (74 mg, 0.091 mmol) were stirred in DMSO (2 mL) under vacuum for 30 min. The flask was flushed with nitrogen and the mixture was heated to 90 0C for 30 min. Upon cooling, the mixture was diluted with methylene chloride and washed with 5% lithium chloride solution (5χ), dried, concentrated, and the residue was purified by column chromatography (silica gel, hexanes/ethyl acetate, 97:3 to 75:25) provide the title compound (337 mg, 62%) as a white solid: 1H NMR (300 MHz, CDCl3) δ 8.96 (s, IH), 8.31 (d, J= 5.4 Hz, IH), 8.06- 8.02 (dd, J= 8.3, 2.1 Hz, IH), 7.87 (d, J= 8.3 Hz, IH), 7.52-7.49 (dd, J= 5.4, 1.4 Hz, IH), 7.36 (s, IH), 3.52 (s, 3H).
b) 4-(5-(Trifluoromethyl)pyridin-2-yl)pyridin-2(l/-/)-one
Chemical Formula: C11H7F3N2O
Exact Mass: 240.05 Molecular Weight: 240.18
Figure imgf000103_0001
[00165] 2'-Methoxy-5-(trifluoromethyl)-2,4'-bipyridine (337 mg, 1.32 mmol) was stirred in concentrated hydrochloric acid (200 mL) at 120 0C for 18 h and then concentrated. The residue was dissolved in MeOH (100 mL) and made basic with 6 N NaOH and re- concentrated until most of the solvent had been removed. The solids were filtered off, washed with water and dried under vacuum to provide the title compound (289 mg, 89%) as a white solid: 1H NMR (300 MHz, DMSO-J6) δ 11.08 (s, IH) 9.10 (s, IH), 8.36-8.33 (dd, J = 8.4, 2.1 Hz, IH), 8.25 (d, J= 8.3 Hz, IH), 7.53 (d, J= 6.8, IH), 7.09 (d, J= 1.3 Hz, IH), 6.90 (dd, J= 6.8, 1.6 Hz, IH).
c) 5-Bromo- 1 -(3-chloropropyl)- lH-indazole
Figure imgf000103_0002
[00166] To a solution of 5-bromo-lH-indazole (3.0 g, 15 mmol) in DMSO (15 niL) was added l-bromo-3-chloropropane (2.0 mL, 20 mmol) and K2CO3 (6.33 g, 45.9 mmol). The reaction was stirred at room temperature for 72 h; then the reaction was diluted with methylene chloride (100 mL) and washed with a 5 % LiCl solution (4x). The organics were washed with brine (50 mL), dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (40 g ISCO column, hexanes/EtOAc, 95:5 to 65:35) gave the title compound (2.14 g, 51%) as a white powder: 1H NMR (500 MHz, CDCl3) δ 7.95 (d, J= 0.5, IH), 7.87 (d, J= 1.5 Hz, IH), 7.48-7.45 (dd, J= 8.9, 1.8 Hz, IH), 7.38 (d, J= 8.9 Hz, IH), 4.54 (t, J= 6.4 Hz, 2H) 3.47 (t, J = 6.0 Hz, 2H) 2.42-2.37 (m, 2H); ESI MS m/z 273 [M + H]+.
d) 5-Bromo- 1 -(3-(pyrrolidin- 1 -yl)propyl)- lH-indazole
Figure imgf000104_0001
[00167] To a solution of 5-bromo-l-(3-chloropropyl)-lH-indazole (2.14 g, 7.84 mmol) in DMF (40 mL) was added potassium iodide (1.3 mg, 7.8 mmol) and K2CO3 (5.40 g, 39.2 mmol) under N2 (g). Then pyrolidine (17.2 ml, 26.6 mmol) was added. The reaction was heated to 50 0C for 20 h. The reaction was diluted with EtOAc (100 mL) and washed with a 5 % LiCl solution (4x). The organics were dried (Na2SO4), filtered, and concentrated to give the title compound (2.48 g, quant.) as a brown oil: 1H NMR (300 MHz, CDCl3) δ 7.93 (s, IH), 7.85 (d, J = 1.2 Hz, IH), 7.45-7.41 (dd, J = 8.9, 1.7 Hz, IH), 7.36 (d, J = 8.9 Hz, IH), 4.45 (t, J= 6.7 Hz, IH), 2.44-2.36 (m, 6H), 2.15-2.07 (m, 2H), 1.78-1.74 (m, 4H); ESI MS m/z 308 [M + H]+.
e) 1 -(I -(3-(Pyrrolidin- 1 -yl)propyl)- lH-indazol-5-yl)-4-(5-(trifluoromethyl)pyridin-2- yl)pyridin-2(lH)-one hydrochloride Chemical Formula: C25H25ClF3N5O
Exact Mass: 503.17 Molecular Weight: 503.95
Figure imgf000105_0001
[00168] 5-Bromo-l-(3-(pyrrolidin-l-yl)propyl)-l/f-indazole (137 mg, 0.444 mmol) and A- (5-(trifluoromethyl)pyridin-2-yl)pyridin-2(l/-/)-one (82 mg, 0.342 mmol) were reacted according to Example 37 (steps e and h) to provide the title compound (57.9 mg, 29%) as a yellow solid: mp 110-120 0C; 1H NMR (500 MHz, CD3OD) δ 9.05 (s, IH), 8.29-8.27 (dd, J = 8.4, 2.1 Hz, IH), 8.22 (d, J= 8.4 Hz, IH), 8.20 (s, IH), 7.90 (d, J= 1.8 Hz, IH), 7.83 (d, J = 7.2 Hz, IH), 7.79 (d, J= 8.9 Hz, IH), 7.53-7.51 (dd, J= 8.9, 1.9 Hz, IH), 7.40 (d, J= 1.7 Hz, IH), 7.27-7.25 (dd, J= 7.2, 1.9 Hz, IH), 4.63 (t, J= 6.5, 2H), 3.67-3.63 (m, 2H), 3.27- 3.24 (m, 2H), 3.08-3.03 (m, 2H), 2.43-2.36 (m, 2H), 2.16-2.12 (m, 2H) 2.03-1.99 (m, 2H); ESI MS m/z 468 [M + H]+; HPLC (Method D) >99% (AUC), tR = 18.0 min.
Example 62
Preparation of 4-(4-(Piperidin- 1 -yPphenyl)- 1 -( 1 -(2-(pyrrolidin- 1 -yPethyD- 1 H-indazol-5- yl)pyridin-2(lH)-one hydrochloride
a) l-(4-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)piperidine
Chemical Formula: C17H26BNO2
Exact Mass: 287.21 Molecular Weight: 287.20
Figure imgf000105_0002
[00169] To a solution of l-(4-bromophenyl)piperidine (250 mg, 1.04 mmol) in DMSO (6 mL) was added δώ-pinacolatodiboron (314 mg, 1.24 mmol) and KOAc (306 mg, 3.12 mmol). The reaction was degassed under vacuum for 30 min. then the flask was flushed N2 (g). PdCl2dppf (85 mg, 0.104 mmol) was added, then the reaction was heated to 60 0C for 20 h. Upon cooling, the reaction was then diluted with methylene chloride (50 rnL) and washed with a 5 % LiCl solution (4x). The organics were dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (12 g ISCO column, methylene chloride/[MeOH/NH4OH 10:1)], 100:0 to 85:15) gave the title compound (262 mg, 86%) as a white powder: ESI MS m/z 288 [M + H]+.
b) 4-(4-(Piperidin- 1 -yl)phenyl)- 1 -(I -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)pyridin- 2(lH)-one hydrochloride
Chemical Formula: C29H34ClN5O
Exact Mass: 503.25 Molecular Weight: 504.07
Figure imgf000106_0001
[00170] Following the procedure in Example 1, but substituting l-(4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)phenyl)piperidine (268 mg, 0.434 mmol) for phenylboronic acid, the title compound (54.5 mg, 23%) was prepared as a orange solid: melting point (mp) 80- 90 0C; 1H NMR (500 MHz, CD3OD) δ 8.26 (s, IH), 7.98 (d, J = 8.7 Hz, 2H), 7.91 (d, J = 1.8 Hz, IH), 7.84-7.76 (m, 4H), 7.55-7.53 (dd, J = 9.0, 1.9 Hz, IH), 6.93 (d, J = 1.9 Hz, IH), 6.87-6.85 (dd, J = 7.2, 2.0 Hz, IH), 4.89 (t, J = 5.7 Hz, 2H), 3.88 (t, J = 5.7 Hz, 2H), 3.72-3.66 (m, 6H), 3.19-3.15 (m, 2H), 2.18-2.16 (m, 2H), 2.06-2.01 (m, 6H), 1.83 (m, 2H); ESI MS m/z 468 [M + H]+; HPLC (Method C) 97.1% (AUC), tR = 10.2 min.
Example 63
Preparation of 4-(5-Methylbenzo[<i]oxazol-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yDethyl)- lH-indazol- 5-yl)pyridin-2(l/-f)-one hydrochloride
a) 5 -Methyl-2-(methylthio)benzo [<i]oxazo Ie Beilstein Registry Number 4849575 Chemical Formula: C9H9NOS
Exact Mass: 179.04
Figure imgf000107_0001
Molecular Weight: 179.24
[00171] To a solution of 5-methylbenzoxazoline-2-thione (1.0 g, 6.2 mmol) in Acetone (50 niL) was added iodomethane (0.6 niL, 9.9 mmol) and K2CO3 (3.4 mg, 24.8 mmol). The flask was flushed with N2 (g), and the mixture was stirred at room temperature for 20 h. The reaction was then filtered through celite, rinsed with acetone, and then the organics were concentrated. The resulting solid was dissolved in EtOAc and washed with water (3χ). The organics were dried (Na2SO4), filtered, and concentrated. Purification by flash chromatography (12 g ISCO column, hexanes/EtOAc, 95:5 to 70:30) gave the title compound (1.04 g, 93%) as a white powder: 1H NMR (300 MHz, CDCl3) δ 7.39 (s, IH), 7.30 (d, J= 8.3 Hz, IH), 7.06-7.02 (dd, J= 8.3, 0.9 Hz, IH), 2.75 (s, 3H), 2.44 (s, 3H); ESI MS m/z 180 [M + H]+.
b) 4-(5-Methylbenzo[<i]oxazol-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Chemical Formula: C26H26ClN5O2
Exact Mass: 475.18 Molecular Weight: 475.97
Figure imgf000107_0002
[00172] 1 -(I -(2-(Pyrrolidin- 1 -yl)ethyl)- lH-indazol-5-yl)-4-(trimethylstannyl)pyridin- 2(lH)-one (prepared according to Example 30, step a) (250 mg, 0.530 mmol), 5-methyl-2- (methylthio)benzo[d]oxazole (198 mg, 1.11 mmol), copper(II) bromide (331 mg, 1.16 mmol) and palladium tetrakistriphenylphosphine (61 mg, 0.053 mmol) were stirred in dry THF (6 mL) under a nitrogen atmosphere. The mixture was heated to 70 0C for 20 h. Upon cooling, the mixture was diluted with THF, filtered through celite and rinsed with THF/methylene chloride. The organics were concentrated and purified by flash chromatography (40 g ISCO column eluting with methylene chloride and methanol/ammonia (10:1); 100% methylene chloride to 15% methanol/ammonia over 35 min at 40 mL/min) then further purified by Preparatory HPLC. The appropriate fractions were concentrated and partitioned between methylene chloride and Na2COs. The organics were dried with Na2SO4, filtered, and concentrated to provide the free base. This was dissolved in methylene chloride (2 mL) and treated with 1 equivalent of 2 M HCl in Et2O, and the mixture was concentrated to provide the title compound (55.8 mg, 24%) as a yellow solid: mp 270-280 0C; 1H NMR (500 MHz, CD3OD) δ 8.27 (s, IH), 7.94 (d, J = 1.7 Hz, IH), 7.88 (d, J= 7.1 Hz, IH), 7.83 (d, J= 8.9 Hz, IH), 7.64-7.61 (m, 2H), 7.58-7.55 (dd, J = 8.9, 1.9 Hz, IH), 7.44 (d, J= 1.7 Hz, IH), 7.37 (d, J= 8.4 Hz, IH), 7.24-7.22 (dd, J= 7.1, 1.9 Hz, IH), 4.89 (t, J = 5.7, 2H), 3.88 (t, J = 5.7, 2H), 3.74-3.70 (m, 2H), 3.21-3.16 (m, 2H), 2.51 (s, 3H), 2.20-2.16 (m, 2H) 2.04-2.01 (m, 2H); ESI MS m/z 440 [M + H]+; HPLC (Method C) 98.7% (AUC), tR = 13.1 min.
Example 64
Preparation of 4-(5-Methoxy- l/-f-indol-2-yl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(l/-/)-one hydrochloride
Chemical Formula: C27H28ClN5O2
Exact Mass: 489.19 Molecular Weight: 490.00
Figure imgf000108_0001
[00173] Following the procedure of Example 1, but substituting 5-methoxyindole-2- boronic acid for phenylboronic acid, the title compound (67 mg, 68%) was prepared as a yellow solid: mp 275-280 0C; 1H NMR (500 MHz, CD3OD) δ 8.25 (s, IH), 7.90 (d, J= 1.6 Hz, IH), 7.80 (d, J = 8.9 Hz, IH), 7.68-7.67 (dd, J = 6.9, 1.2 Hz, IH), 7.55-7.52 (dd, J = 8.8, 1.9 Hz, IH), 7.33 (d, J= 8.9 Hz, IH), 7.08 (d, J= 2.2 Hz, IH), 7.06 (s, IH), 6.98-6.95 (m, 2H), 6.90-6.88 (dd, J = 8.9, 2.5 Hz, IH), 4.88 (t, J = 5.8 Hz, 2H), 3.87 (t, J = 5.8 Hz, 2H), 3.83 (s, 3H), 3.77-3.67 (br m, 2H), 3.22-3.12 (br m, 2H), 2.20-2.11 (br m, 2H), 2.08- 1.97 (br m, 2H); ESI MS m/z 454 [M + H]+; HPLC (Method C) >99% (AUC), tR = 12.4 min. Example 65
Preparation of 4-(5-Methoxy- 1 -methyl- lH-indol-2-vfl- 1 -(I -(2-(pyrroridin- 1 -vDethvO- IH- indazol-5-yl)pyridin-2(lH)-one dihydrochloride
Chemical Formula: C28H31Cl2N5O2
Exact Mass: 539.19 Molecular Weight: 540.48
Figure imgf000109_0001
[00174] Under a nitrogen atmosphere 4-(5-methoxy-lH-indol-2-yl)-l-(l-(2-(pyrrolidin-l- yl)ethyl)-lH-indazol-5-yl)pyridin-2(lH)-one (47 mg, 0.10 mmol) was dissolved in DMF (5 mL) and NaH (60% dispersion, 5 mg, 0.12 mmol) was added. After 20 minutes MeI (21 mg, 9.3 μL, 0.15 mmol) was added and the mixture was stirred overnight. The solid was filtered off, purified by prep HPLC and converted to the dihydrochloride as in Example 1 to provide the title compound (10 mg, 18%) as a yellow solid: 1H NMR (500 MHz, CD3OD) δ 8.27 (s, IH), 7.92 (d, J = 1.7 Hz, IH), 7.82 (d, J = 8.8 Hz, IH), 7.75 (d, J = 6.6 Hz, IH), 7.57-7.55 (dd, J= 8.9, 1.9 Hz, IH), 7.37 (d, J= 8.9 Hz, IH), 7.11 (d, J= 2.4 Hz, IH), 6.96- 6.94 (dd, J= 8.9, 2.4 Hz, IH), 6.80-6.78 (m, 3H), 4.89 (t, J= 5.7 Hz, 2H), 3.88 (s, 3H), 3.87 (t, J= 5.7 Hz, 2H), 3.83 (s, 3H), 3.72 (br m, 2H), 3.18 (br m, 2H), 2.17 (br m, 2H), 2.03 (br m, 2H); ESI MS m/z 468 [M + H]+; HPLC (Method C) 98.8% (AUC), tR = 12.8 min.
Example 66
Preparation of 4-(4-(lH-Pyrazol- 1 -vOphenylV 1 -(I -(2-(pyrroridin- 1 -vDethvO- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Figure imgf000110_0001
[00175] Following the procedure of Example 1, but substituting 4-(lH-pyrazol-l- yl)phenylboronic acid for phenylboronic acid, the title compound (64 mg, 60%) was prepared as a brown solid: mp 271-275 0C; 1H NMR (500 MHz, CD3OD) δ 8.34 (d, J= 2.5 Hz, IH), 8.26 (s, IH), 7.95-7.90 (m, 5H), 7.83 (d, J= 8.9 Hz, IH), 7.78-7.76 (m, 2H), 7.56- 7.54 (d, J= 8.9, 1.9 Hz, IH), 6.95 (d, J= 1.9 Hz, IH), 6.90 (dd, J= 7.1, 2.0 Hz, IH), 6.58 (t, J = 1.9 Hz, IH), 4.89 (t, J = 5.8 Hz, 2H), 3.89 (t, J = 5.8 Hz, 2H), 3.76-3.68 (br m, 2H), 3.21-3.13 (br m, 2H), 2.22-2.12 (br m, 2H), 2.08-1.96 (br m, 2H); ESI MS m/z 451 [M + H]+; HPLC (Method C) 95.1% (AUC), tR = 11.7 min.
Example 67
Preparation of 4-(4-Methylcyclohex- 1 -enyP- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(l/-/)-one hydrochloride
Figure imgf000110_0002
[00176] Following the procedure of Example 1, but substituting 4-methylcyclohex-l- enylboronic acid for phenylboronic acid, the title compound (76 mg, 54%) was prepared as an orange solid: mp 256-260 0C; 1H NMR (500 MHz, CD3OD) δ 8.23 (s, IH), 7.84 (d, J = 1.4 Hz, IH), 7.78 (d, J = 8.9 Hz, IH), 7.55 (d, J = 7.2 Hz, IH), 7.49-7.47 (dd, J = 8.9, 1.9 Hz, IH), 6.70-6.68 (dd, J = 7.3, 1.9 Hz, IH), 6.57-6.54 (m, 2H), 4.87 (t, J = 5.7 Hz, 2H), 3.86 (t, J = 5.7 Hz, 2H), 3.73-3.69 (m, 2H), 3.20-3.13 (m, 2H), 2.46-2.36 (m, 3H), 2.21- 2.12 (m, 2H), 2.05-1.97 (m, 2H), 1.96-1.85 (m, 2H), 1.79-1.71 (m, IH), 1.44-1.34 (m, IH), 1.03 (d, J = 6.6 Hz, 3H); ESI MS m/z 403 [M + H]+; HPLC (Method C) >99% (AUC), tR =13.4 min.
Example 68
Preparation of 4-(4-Methylcyclohexyl)- 1 -(I -(2-(pyrrolidin- 1 -yPethyl)- lH-indazol-5- yl)pyridin-2(lH)-one hydrochloride
Chemical Formula: C25H33ClN4O
Exact Mass: 440.23 Molecular Weight: 441.01
Figure imgf000111_0001
[00177] 4-(4-Methylcyclohex- 1 -enyl)- 1 -(I -(2-(pyrrolidin- 1 -yl)ethyl)- lH-indazol-5- yl)pyridin-2(l/-/)-one (50 mg, 0.12mmol) was reacted following the procedure of Example 1 (step d) to provide the title compound (mixture of E and Z isomers) (31 mg, 60%) as a white solid: mp 80-85 0C; 1H NMR (500 MHz, CD3OD) δ 8.21 (s, IH), 7.82 (s, IH), 7.77 (d, J = 8.8 Hz, IH), 7.57-7.56 (m, IH), 7.47-7.44 (m, IH), 6.54-6.45 (m, 2H), 4.82 (m, 2H), 3.71- 3.64 (br m, 2H), 3.29-3.28 (br m, 3H), 2.61-2.55 (t, J= 8.5, 0.6 Hz, 0.6H), 2.48-2.41 (t, J = 13.2 Hz, 0.4H), 2.02 (s, 4H), 1.97-1.67 (m, 6H), 1.59-1.42 (m, 3H), 1.34 (q, J = 13.1 Hz, IH), 1.05 (d, J = 7.3 Hz, 2H), 0.92 (d, J = 7.0 Hz, IH); ESI MS m/z 405 [M + H]+; HPLC (Method C) 98.6% (AUC), tR = 13.4 min.
Binding Assay I for Human Melanin-Concentrating Hormone (MCHi) receptor
[00178] Evaluation of the affinity of compounds for the human MCHi receptor was accomplished in transfected Chinese Hamster Ovary (CHO) cells determined in a radioligand binding assay, as described in MacDonald et al, "Molecular characterization of the melanin-concentrating hormone/receptor complex: identification of critical residues involved in binding and activation", MoI Pharmacol, 58.:217 (2000). Cell membrane homogenates (5 μg protein) were incubated for 60 min at 22°C with 0.1 nM [125I][Phe13,Tyr19]-MCH in the absence or presence of the test compound in a buffer containing 25 mM Hepes/Tris (pH 7.4), 5 mM MgCl2, 1 niM CaCl2 and 0.5% bovine serum albumin (BSA). Nonspecific binding was determined in the presence of 0.1 μM MCH. Following incubation, the samples were filtered rapidly under vacuum through glass fiber filters (GF/B, Packard) and rinsed several times with an ice-cold buffer containing 25 mM Hepes/Tris (pH 7.4), 500 mM NaCl, 5 mM MgCl2, 1 mM CaCl2 and 0.1% BSA using a 96- sample cell harvester (Unifilter, Packard). The filters were dried, then counted for radioactivity in a scintillation counter (Topcount, Packard) using a scintillation cocktail (Microscint 0, Packard).
[00179] The results are expressed as a percent inhibition of the control radioligand specific binding. The IC50 value (concentration causing a half-maximal inhibition of control specific binding) and Hill coefficient (TIH) were determined by non- linear regression analysis of the competition curve using Hill equation curve fitting. The inhibition constant (K1) was calculated from the Cheng Prusoff equation:
(K1 = IC50/(1+(L/KD)), where L = concentration of radioligand in the assay, and KD = affinity of the radioligand for the receptor).
Binding Assay II for Human Melanin-Concentrating Hormone (MCHi) receptor
[00180] Evaluation of the affinity of compounds for the human MCHi receptor was accomplished using tri-[ H]-labeled 4-(benzyloxy)-l-(l-(2-(pyrrolidin-l-yl)ethyl)-lH- indazol-5-yl)pyridin-2(lH)-one and membranes prepared from stable CHO-Kl cells expressing the human MCHi receptor obtained from Euroscreen (Batch 1138). Cell membrane homogenates (8.92 μg protein) were incubated for 60 min at 25°C with 1.4 nM of the [3H]-labeled compound in the absence or presence of the test compound in 50 mM Tris- HCl buffer, pH 7.4. Nonspecific binding was determined in the presence of 50 μM l-(5-(4- cyanophenyl)bicyclo[3.1.0]hexan-2-yl)-3-(4-fluoro-3-(trifluoromethyl)phenyl)- 1 -(3-(4- methylpiperazin-l-yl)propyl)urea. Following incubation, the samples were filtered rapidly under vacuum through Skatron 11731 filters, pre-soaked in 0.5% polyethylenimine, and washed with ice-cold 50 niM Tris-HCl buffer, pH 7.4, (wash setting 9,9,0) using a Skatron cell harvester. The filters were counted for radioactivity in a liquid scintillation counter (Tri-Carb 2100TR, Packard) using a scintillation cocktail (Ultima Gold MV, Perkin Elmer).
[00181] The results are expressed as a percent inhibition of the control radioligand specific binding. The IC50 value (concentration causing a half-maximal inhibition of control specific binding) and Hill coefficient (TIH) were determined by non- linear regression analysis of the competition curve using Hill equation curve fitting. The inhibition constant (K1) was calculated from the Cheng Prusoff equation: (K1 = IC50/(1+(L/KD)), where L = concentration of radioligand in the assay, and KD = affinity of the radioligand for the receptor.
[00182] By methods described above, the compounds listed in Table 1 were synthesized and tested for biological activity. All of the compounds in Table 1 exhibited K1 of less than or equal to 3.5 μM in the MCHi binding assay I or II.
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000122_0001
Figure imgf000123_0001
Figure imgf000124_0001
Figure imgf000125_0001
Figure imgf000126_0001
inhibition.
[00183] The present invention is not limited to the compounds found in the above examples, and many other compounds falling within the scope of the invention may also be prepared using the procedures set forth in the above synthetic schemes. The preparation of additional compounds of formula I using these methods will be apparent to one of ordinary skill in the chemical arts.
[00184] The invention has been described in detail with particular reference to some embodiments thereof, but it will be understood by those skilled in the art that variations and modifications can be effected within the spirit and scope of the invention.

Claims

CLAIMS What is claimed is:
1. A compound of formula I:
Figure imgf000128_0001
wherein n is 0 or 1 ;
R is NR1R2, wherein R1 and R2 are each independently H and optionally substituted alkyl, or R1 and R2, together with the N atom to which they are attached, form a 4-7 membered optionally substituted non-aromatic heterocyclic ring which optionally contains 1 or 2 heteroatoms in addition to the N atom shown; R3 and R4 are each independently H or alkyl; or R may combine with either R3 or R4 to form an optionally substituted pyrrolidin-2-yl; B is selected from ary, heteroaryl, and cycloalkyl; R5, R6, R7 are each independently selected from H, -OH, -O-alkyl, alkyl, halo, -CF3, and
-CN, -O-aryl, heteroaryl, heterocyclyl; and R14 is H or -OH; and provided that when n is 0, R3 and R4 are H and R is pyrrolidin-1-yl, then
(a) when B is a 5-7-membered monocyclic aromatic heterocycle, at least one heteroatom of said monocyclic aromatic heterocycle is adjacent to the position where B attaches to the pyridinone moiety, and
(b) when B is phenyl which is substituted at the 2-position by methoxy or at the 3 -position by methyl, there is at least one additional substituent on the phenyl ring.
2. A compound according to claim 1, with the further proviso that when n is 0, R3 and R4 are H, R is pyrrolidin-1-yl and B is a bicyclic aromatic heterocycle, then either
(i) at least one heteroatom of said bicyclic aromatic heterocycle is adjacent to the position where B attaches to the pyridinone moiety, or (ii) the ring of said bicyclic aromatic heterocycle which attaches to the pyridinone moiety does not contain a heteratom.
3. A compound according to claim 1 or 2, wherein R is selected from the group consisting of pyrrolidin-1-yl, 3-hydroxypyrrolidin-l-yl, morpholin-4-yl, 3- hydroxymethylpyrrolidin-1-yl, dimethylamino, piperazin-1-yl, amino, and 2-oxa-5- azabicyclo[2.2.1]heptane-5-yl.
4. A compound according to any of claims 1 to 3, wherein R is selected from S-3- hydroxypyrrolidin- 1 -yl, R-3 -hydroxypyrrolidin- 1 -yl, S-3 -hydroxymethylpyrro lidin- 1 - yl and R-3 -hydroxymethylpyrro lidin- 1-yl, and (lS,4S)-2-oxa-5- azabicyclo[2.2.1]heptane-5-yl.
5. A compound according to any of claims 1 to 4, wherein R3 and R4 are both H.
6. A compound according claim 1, wherein R is combined with either R3 or R4 to form an optionally substituted pyrrolidin-2-yl
7. A compound according to claim 1, wherein B, taken together with R5, R6 and R7, is 4-trifluoromethylphenyl and n is 1.
8. A compound according to claim 7, wherein R14 is -OH.
9. A compound according to any of claims 1 to 7, wherein n is 0.
10. A compound according to any of claims to 7, wherein n is 1.
11. A compound according to any of claims 1 to 10, wherein B is phenyl.
12. A compound according to claim 11, wherein B, taken together with R5, R6 and R7, is selected from the group consisting of phenyl, 4-chlorophenyl, 4-fluorophenyl, 3- chlorophenyl, 3-phenoxyphenyl, 4-trifluoromethylphenyl, 3-trifluoromethylphenyl, 2,4-dichlorophenyl, 4-methylphenyl, 4-trifluoromethoxyphenyl, 4-cyanophenyl, 4- methoxyphenyl, 2-cyano-4-fluorophenyl, 2,4-dimethoxyphenyl, 2,4-difluorophenyl, 4-isopropoxyphenyl, 2,4-di-trifluoromethylphenyl, 4-/?-butoxy-2-methylphenyl, 2- methylphenyl, 4-benzyloxy-2-methylphenyl, 4-chloro-2-methoxyphenyl, benzodioxol-5-yl, 4-methoxy-2-methylphenyl and 2-chloro-4-trifluoromethylphenyl, 4-chloro-2-fluorophenyl, 4-triflluoromethyl-2-fluorophenyl, 4-methoxy-2- fluorophenyl, 4-methoxy-2-chlorophenyl, 4-ethoxyphenyl, 4-trifluoromethoxy-2- fluorophenyl, and 4-trifluoromethoxy-2-methylphenyl, 4-(piperidine-l-yl)phenyl, and 4-( lH-pyrazol- 1 -yl)phenyl.
13. A compound according to any of claims 1 to 10, wherein B, taken together with R5, R6 and R7, is selected from napthalen-1-yl and naphthalen-2-yl.
14. A compound according to any of claim 1 to 10, wherein B, taken together with R5, R6 and R7, is benzothiophene or benzofuran.
15. A compound according to claim 14, wherein B, taken together with R5, R6 and R7, is selected from benzothiophene-2-yl and benzofuran-2-yl.
16. A compound according to any of claims 1 to 10, wherein B is indole.
17. A compound according to claim 16, wherein B, taken together with R5, R6 and R7, is 5 -methoxyindo 1-2-yl.
18. A compound according to claim 16, wherein B is N-alkyl substituted indole.
19. A compound according to claim 17, wherein B, taken together with R5, R6 and R7, is selected from l-methylindol-2-yl, l-methylindol-5-yl, and l-methyl-5-methoxyindol- 2-yl.
20. A compound according to any of claims to 10, wherein B is pyridine.
21. A compound according to claim 20, wherein B, taken together with R5, R6 and R7, is selected from 5-methylpyridine-2-yl, 5-chloropyridin-2-yl, and 5- trifluoromethylpyridin-2-yl.
22. A compound according to any of claims 1 to 10, wherein B, taken together with R5, R6 and R7, is selected from quinoline, quinazoline, and optionally substituted pyridazine.
23. A compound according to claim 22, wherein B, taken together with R5, R6 and R7, is selected from quinolin-2-yl, quinazolin-2-yl, and 6-trifluoromethylpyridazin-3-yl.
24. A compound according to any of claims 1 to 10, wherein B, taken together with R5, R6 and R7, is selected from l-methylindazol-5-yl.
25. A compound according to any of claims 1 to 10, wherein B, taken together with R5, R6 and R7, is selected from 5-methylbenzisoxazol-2-yl.
26. A compound according to any of claim 1 to 10, wherein B, taken together with R5, R6 and R7, is selected from 4-methylcyclohex-l-enyl.
27. A compound according to any of claims 1 to 10, wherein B, taken together with R5, R6 and R7, is selected from 4-methylcyclohex-l-yl.
28. A compound according to any of claims 1, wherein the compound is selected from the group comprising:
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
29. A compound according to claim 28, which is a pharmaceutically acceptable salt thereof.
30. A compound according to claim 1, wherein the compound is selected from the group comprising:
Figure imgf000136_0002
Figure imgf000136_0003
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000138_0002
Figure imgf000138_0003
Figure imgf000139_0001
and H3C
31. A compound according to claim 30, which is a pharmaceutically acceptable salt thereof.
32. A compound according to claim 1, wherein the compound is selected from the group comprising:
Figure imgf000139_0002
Figure imgf000139_0003
Figure imgf000140_0001
Figure imgf000140_0002
Figure imgf000141_0001
33. A compound according to claim 32, which is a pharmaceutically acceptable salt thereof.
34. A compound according to claim 1, wherein the compound is selected from the group comprising:
Figure imgf000141_0002
Figure imgf000142_0001
Figure imgf000142_0002
Figure imgf000143_0001
35. A compound according to claim 34, which is a pharmaceutically acceptable salt thereof.
36. A pharmaceutical composition comprising a compound according to any of claims 1 to 35 and a pharmaceutically acceptable carrier, excipient or diluent therefor.
37. A method of treating non-alcoholic fatty liver disease, comprising administering to a patient in need of such treatment a therapeutically effective amount of a compound according to any of claims 1 to 35.
38. A method of treating obesity, comprising administering to a patient in need of obesity reduction an obesity-reducing effective amount of a compound according to any claims 1 to 35.
39. A method of treating anxiety, comprising administering to a patient in need of such treatment an effective amount of a compound according to any of claims 1 to 35.
40. A method of treating depression, comprising administering to a patient in need of such treatment an effective amount of a compound according to any of claims 1 to
35.
41. A method of treating a disease or condition which is susceptible to treatment with an MCHi receptor modulator, comprising administering to a patient in need thereof a therapeutically effective amount of a compound according to any of claims 1 to 35.
PCT/US2008/070535 2007-07-21 2008-07-18 5-pyridinone substituted indazoles WO2009015037A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
CN200880107571A CN101861311A (en) 2007-07-21 2008-07-18 The indazole that the 5-pyridone replaces
NZ582586A NZ582586A (en) 2007-07-21 2008-07-18 5-pyridinone substituted indazoles
AT08796320T ATE544759T1 (en) 2007-07-21 2008-07-18 5-PYRIDINONE-SUBSTITUTED INDAZOLES AND PHARMACEUTICAL COMPOSITIONS THEREOF
BRPI0814772-8A2A BRPI0814772A2 (en) 2007-07-21 2008-07-18 INDAZOES REPLACED BY 5-PYRIDINONE
JP2010518311A JP2010534248A (en) 2007-07-21 2008-07-18 5-pyridinone substituted indazole
CA2693377A CA2693377A1 (en) 2007-07-21 2008-07-18 5-pyridinone substituted indazoles
AU2008279321A AU2008279321B2 (en) 2007-07-21 2008-07-18 5-pyridinone substituted indazoles
EP08796320A EP2176251B1 (en) 2007-07-21 2008-07-18 5-pyridinone substituted indazoles and pharmaceutical compositions thereof
ES08796320T ES2382982T3 (en) 2007-07-21 2008-07-18 Indazoles substituted with 5-pyridinone and pharmaceutical compositions thereof
HK10109173.8A HK1142608A1 (en) 2007-07-21 2010-09-24 5-pyridinone substituted indazoles and pharmaceutical compositions thereof 5-

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US95120107P 2007-07-21 2007-07-21
US60/951,201 2007-07-21

Publications (2)

Publication Number Publication Date
WO2009015037A2 true WO2009015037A2 (en) 2009-01-29
WO2009015037A3 WO2009015037A3 (en) 2009-04-30

Family

ID=40282093

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/070535 WO2009015037A2 (en) 2007-07-21 2008-07-18 5-pyridinone substituted indazoles

Country Status (13)

Country Link
US (1) US8273770B2 (en)
EP (1) EP2176251B1 (en)
JP (1) JP2010534248A (en)
KR (1) KR20100044225A (en)
CN (1) CN101861311A (en)
AT (1) ATE544759T1 (en)
AU (1) AU2008279321B2 (en)
BR (1) BRPI0814772A2 (en)
CA (1) CA2693377A1 (en)
ES (1) ES2382982T3 (en)
HK (1) HK1142608A1 (en)
NZ (1) NZ582586A (en)
WO (1) WO2009015037A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8268868B2 (en) 2007-01-10 2012-09-18 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles
US8273770B2 (en) 2007-07-21 2012-09-25 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles
WO2019037640A1 (en) * 2017-08-22 2019-02-28 Js Innopharm (Shanghai) Ltd Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
CN112876411A (en) * 2019-02-21 2021-06-01 杭州阿诺生物医药科技有限公司 Compound and application thereof in synthesis of PDL1 antagonist drug molecules

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5501251B2 (en) 2008-01-11 2014-05-21 アルバニー モレキュラー リサーチ, インコーポレイテッド (1-Azinone) substituted pyridoindoles as MCH antagonists
WO2011003012A1 (en) 2009-07-01 2011-01-06 Albany Molecular Research, Inc. Azinone-substituted azapolycycle mch-1 antagonists, methods of making, and use thereof
EP2448585B1 (en) * 2009-07-01 2014-01-01 Albany Molecular Research, Inc. Azinone-substituted azepino[b]indole and pyrido-pyrrolo-azepine mch-1 antagonists, methods of making, and use thereof
WO2011003021A1 (en) 2009-07-01 2011-01-06 Albany Molecular Research, Inc. Azinone-substituted azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine mch-1 antagonists, methods of making, and use thereof
US8629158B2 (en) 2009-07-01 2014-01-14 Albany Molecular Research, Inc. Azabicycloalkane-indole and azabicycloalkane-pyrrolo-pyridine MCH-1 antagonists, methods of making, and use thereof
US8993765B2 (en) 2010-12-21 2015-03-31 Albany Molecular Research, Inc. Tetrahydro-azacarboline MCH-1 antagonists, methods of making, and uses thereof
WO2012088038A2 (en) 2010-12-21 2012-06-28 Albany Molecular Research, Inc. Piperazinone-substituted tetrahydro-carboline mch-1 antagonists, methods of making, and uses thereof
CA2850570A1 (en) * 2011-09-30 2013-04-04 Glaxosmithkline Llc Methods of treating cancer
CN107011283A (en) * 2017-05-31 2017-08-04 湖南华腾制药有限公司 A kind of methyl mercapto replaces the preparation method of benzo [d] oxazole derivatives

Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1A (en) 1836-07-13 John Ruggles Locomotive steam-engine for rail and other roads
WO1990009781A1 (en) 1989-02-23 1990-09-07 Rorer International (Holdings), Inc. Therapeutic aerosol formulations
WO1991004011A1 (en) 1989-09-20 1991-04-04 Riker Laboratories, Inc. Medicinal aerosol formulations
US5230884A (en) 1990-09-11 1993-07-27 University Of Wales College Of Cardiff Aerosol formulations including proteins and peptides solubilized in reverse micelles and process for making the aerosol formulations
US5292499A (en) 1990-09-11 1994-03-08 University Of Wales College Of Cardiff Method of preparing medical aerosol formulations including drug dissolved in reverse micelles
EP0736299A1 (en) 1995-04-03 1996-10-09 Cerbios-Pharma S.A. Process for preparing a liposomal, water-dispersable, solid, dry, therapeutic formulation for oral administration
WO1996040089A2 (en) 1995-06-07 1996-12-19 Abbott Laboratories Aerosol drug formulations containing vegetable oils
WO1997011682A2 (en) 1995-09-26 1997-04-03 University Of Pittsburgh Emulsion and micellar formulations for the delivery of biologically active substances to cells
WO1997013500A2 (en) 1995-10-12 1997-04-17 Supergen, Inc. LIPOSOME FORMULATIONS OF 5β STEROIDS
WO1999059550A1 (en) 1998-05-20 1999-11-25 The Liposome Company, Inc. Novel particulate formulations
WO2000000176A1 (en) 1998-06-30 2000-01-06 Quadrant Healthcare (Uk) Limited Microparticle formulation for inhalation
WO2001013891A2 (en) 1999-08-25 2001-03-01 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
WO2001060341A1 (en) 2000-02-17 2001-08-23 Rijksuniversiteit Groningen Powder formulation
US6290987B1 (en) 1998-09-27 2001-09-18 Generex Pharmaceuticals, Inc. Mixed liposome pharmaceutical formulation with amphiphiles and phospholipids
US6294153B1 (en) 1998-12-21 2001-09-25 Generex Pharmaceuticals, Inc. Aerosol pharmaceutical formulation for pulmonary and nasal delivery
WO2001078694A2 (en) 2000-04-17 2001-10-25 Vectura Limited Formulations for use in inhaler devices
WO2001078696A2 (en) 2000-04-17 2001-10-25 Vectura Limited Formulations for use in inhaler devices
US6309671B1 (en) 1995-04-14 2001-10-30 Inhale Therapeutic Systems Stable glassy state powder formulations
US20010036481A1 (en) 1999-08-25 2001-11-01 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
US20010038824A1 (en) 2000-01-20 2001-11-08 Ikuo Horii Intranasal cyclic peptide formulations
US20020061336A1 (en) 1999-02-03 2002-05-23 O'connor Barbara Horsey Hydrogel particle formulation
US6436367B1 (en) 1998-12-21 2002-08-20 Generex Pharmaceuticals Inc. Aerosol formulations for buccal and pulmonary application
WO2002080884A2 (en) 2001-03-21 2002-10-17 Boehringer Ingelheim Pharma Gmbh & Co. Kg Powder inhaler formulations
US20030008013A1 (en) 1998-06-30 2003-01-09 Stuart Robinson Formulation for inhalation
US20030019437A1 (en) 2001-07-26 2003-01-30 John Fore Ungulate game animal feed system and method
US6524557B1 (en) 1994-12-22 2003-02-25 Astrazeneca Ab Aerosol formulations of peptides and proteins
WO2003015750A1 (en) 2001-08-16 2003-02-27 Baxter International, Inc. Propellant-based microparticle formulations
EP1338272A1 (en) 1998-12-21 2003-08-27 Generex Pharmaceuticals Inc. Aerosol formulations for buccal and pulmonary application comprising chenodeoxycholate or deoxycholate
WO2003072080A1 (en) 2002-02-22 2003-09-04 Advanced Inhalation Research, Inc. Inhalable formulations for sustained release
US20030165436A1 (en) 2000-04-17 2003-09-04 Staniforth John Nicholas Formulations for use in inhaler devices
WO2003079885A2 (en) 2002-03-20 2003-10-02 Advanced Inhalation Research, Inc. Inhalable sustained therapeutic formulations
US6632456B1 (en) 1993-06-24 2003-10-14 Astrazeneca Ab Compositions for inhalation
WO2003094886A2 (en) 2002-05-07 2003-11-20 Ferring Bv Desmopressin in an orodispersible dosage form
WO2005018557A2 (en) 2003-08-13 2005-03-03 Pharmacia Corporation Substituted pyridinones
EP1741703A1 (en) 2004-03-05 2007-01-10 Banyu Pharmaceutical Co., Ltd. Pyridone derivative
WO2007029847A1 (en) 2005-09-07 2007-03-15 Banyu Pharmaceutical Co., Ltd. Bicyclic aromatic substituted pyridone derivative
WO2008086404A1 (en) 2007-01-10 2008-07-17 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5225565A (en) * 1988-09-15 1993-07-06 The Upjohn Company Antibacterial 3-(fused-ring substituted)phenyl-5β-amidomethyloxazolidin-2-ones
US5182403A (en) * 1988-09-15 1993-01-26 The Upjohn Company Substituted 3(5'indazolyl) oxazolidin-2-ones
US5393735A (en) * 1990-08-09 1995-02-28 Rohm And Haas Company Herbicidal glutarimides
JPH03253852A (en) 1990-03-05 1991-11-12 Fuji Photo Film Co Ltd Method for processing silver halide photographic sensitive material and final processing composition
DE4017211A1 (en) * 1990-05-29 1991-12-05 Merck Patent Gmbh OXAZOLIDINONE
DE4018830A1 (en) 1990-06-12 1991-12-19 Langhals Heinz New perylene dyestuffs synthesised from perylene blue - used as biochemical tracers, dyestuffs and pigments, in optical light gathering systems, in polymerisation reactions, etc.
DE4338784A1 (en) 1993-11-12 1995-05-18 Langhals Heinz Perylene-3,4-di:carboximide(s) and reaction prods. useful as fluorescent dye
TW279860B (en) * 1993-11-12 1996-07-01 Ciba Geigy Ag
WO1997008150A1 (en) 1995-08-22 1997-03-06 The Du Pont Merck Pharmaceutical Company Substituted cyclic ureas and derivatives thereof useful as retroviral protease inhibitors
ZA966885B (en) * 1995-08-22 1998-02-16 Du Pont Merck Pharma Substituted cyclic ureas and derivatives thereof useful as retroviral protease inhibitors.
AU7257096A (en) 1995-10-04 1997-04-28 Fmc Corporation Herbicidal 6-heterocyclic indazole derivatives
US6107300A (en) * 1996-03-27 2000-08-22 Dupont Pharmaceuticals Arylamino fused pyrimidines
DE19651712A1 (en) 1996-12-12 1998-06-18 Langhals Heinz New pyrrolo: or thieno:perylene-imide compounds
DE10104279A1 (en) 2001-01-31 2002-08-01 Heinz Langhals Fluorescent detection of prions, useful for diagnosis, by binding to fluorescently labeled Congo Red derivative, also treatment of prion diseases e.g. Creutzfeld-Jakob disease
CZ20033053A3 (en) * 2001-05-14 2004-05-12 Bristol@Myersásquibbápharmaácompany Substituted pyrazinones, pyridines and pyrimidines as corticotropin releasing factor ligands
US20030114448A1 (en) * 2001-05-31 2003-06-19 Millennium Pharmaceuticals, Inc. Inhibitors of factor Xa
AU2002326948A1 (en) 2001-09-18 2003-04-01 Bristol-Myers Squibb Company Piperizinones as modulators of chemokine receptor activity
GB0124627D0 (en) 2001-10-15 2001-12-05 Smithkline Beecham Plc Novel compounds
TW200302225A (en) * 2001-12-04 2003-08-01 Bristol Myers Squibb Co Substituted amino methyl factor Xa inhibitors
AU2003277285B2 (en) 2002-10-04 2007-12-13 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
CA2521832A1 (en) 2003-04-11 2004-10-28 Smithkline Beecham Corporation Heterocyclic mchr1 antagonists
ATE532781T1 (en) 2003-06-19 2011-11-15 Glaxosmithkline Llc 5-(ACYLAMINO)INDAZOLE DERIVATIVES AS KINASE INHIBITORS
US7390820B2 (en) * 2003-08-25 2008-06-24 Amgen Inc. Substituted quinolinone derivatives and methods of use
US7320992B2 (en) * 2003-08-25 2008-01-22 Amgen Inc. Substituted 2,3-dihydro-1h-isoindol-1-one derivatives and methods of use
CN1871242A (en) 2003-10-23 2006-11-29 葛兰素集团有限公司 3-(4-aminophenyl) thienopyrimid-4-one derivatives as MCH R1 antagonists for the treatment of obesity, diabetes, depression and anxiety
US7049307B2 (en) * 2003-12-23 2006-05-23 Abbott Laboratories Antagonists of melanin concentrating hormone effects on the melanin concentrating hormone receptor
US20050137243A1 (en) * 2003-12-23 2005-06-23 Souers Andrew J. Antagonists of melanin concentrating hormone effects on the melanin concentrating hormone receptor
US7071182B2 (en) * 2003-12-23 2006-07-04 Abbott Laboratories Antagonists of melanin concentrating hormone effects on the melanin concentrating hormone receptor
AU2005243147A1 (en) * 2004-05-07 2005-11-24 Memory Pharmaceuticals Corporation 1H-indazoles, benzothiazoles, 1,2-benzoisoxazoles, 1,2-benzoisothiazoles, and chromones and preparation and uses thereof
WO2006017257A2 (en) 2004-07-12 2006-02-16 Phenomix Corporation Azetidinone derivatives
BRPI0806506A2 (en) * 2007-01-10 2014-04-22 Albany Molecular Res Inc INDAZOES REPLACED BY 5-FUROPYRIDINONE
WO2009015037A2 (en) 2007-07-21 2009-01-29 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles

Patent Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1A (en) 1836-07-13 John Ruggles Locomotive steam-engine for rail and other roads
WO1990009781A1 (en) 1989-02-23 1990-09-07 Rorer International (Holdings), Inc. Therapeutic aerosol formulations
WO1991004011A1 (en) 1989-09-20 1991-04-04 Riker Laboratories, Inc. Medicinal aerosol formulations
US5348730A (en) 1989-09-20 1994-09-20 Minnesota Mining And Manufacturing Company Method for preparing medicinal aerosol formulation containing coated medicament
US5230884A (en) 1990-09-11 1993-07-27 University Of Wales College Of Cardiff Aerosol formulations including proteins and peptides solubilized in reverse micelles and process for making the aerosol formulations
US5292499A (en) 1990-09-11 1994-03-08 University Of Wales College Of Cardiff Method of preparing medical aerosol formulations including drug dissolved in reverse micelles
US6632456B1 (en) 1993-06-24 2003-10-14 Astrazeneca Ab Compositions for inhalation
US6524557B1 (en) 1994-12-22 2003-02-25 Astrazeneca Ab Aerosol formulations of peptides and proteins
EP0736299A1 (en) 1995-04-03 1996-10-09 Cerbios-Pharma S.A. Process for preparing a liposomal, water-dispersable, solid, dry, therapeutic formulation for oral administration
US6309671B1 (en) 1995-04-14 2001-10-30 Inhale Therapeutic Systems Stable glassy state powder formulations
US20020141945A1 (en) 1995-04-14 2002-10-03 Foster Linda C. Stable glassy state powder formulations
WO1996040089A2 (en) 1995-06-07 1996-12-19 Abbott Laboratories Aerosol drug formulations containing vegetable oils
WO1997011682A2 (en) 1995-09-26 1997-04-03 University Of Pittsburgh Emulsion and micellar formulations for the delivery of biologically active substances to cells
WO1997013500A2 (en) 1995-10-12 1997-04-17 Supergen, Inc. LIPOSOME FORMULATIONS OF 5β STEROIDS
WO1999059550A1 (en) 1998-05-20 1999-11-25 The Liposome Company, Inc. Novel particulate formulations
WO2000000176A1 (en) 1998-06-30 2000-01-06 Quadrant Healthcare (Uk) Limited Microparticle formulation for inhalation
US20030008013A1 (en) 1998-06-30 2003-01-09 Stuart Robinson Formulation for inhalation
US6290987B1 (en) 1998-09-27 2001-09-18 Generex Pharmaceuticals, Inc. Mixed liposome pharmaceutical formulation with amphiphiles and phospholipids
US6436367B1 (en) 1998-12-21 2002-08-20 Generex Pharmaceuticals Inc. Aerosol formulations for buccal and pulmonary application
US6294153B1 (en) 1998-12-21 2001-09-25 Generex Pharmaceuticals, Inc. Aerosol pharmaceutical formulation for pulmonary and nasal delivery
EP1338272A1 (en) 1998-12-21 2003-08-27 Generex Pharmaceuticals Inc. Aerosol formulations for buccal and pulmonary application comprising chenodeoxycholate or deoxycholate
US20020061336A1 (en) 1999-02-03 2002-05-23 O'connor Barbara Horsey Hydrogel particle formulation
US20040018243A1 (en) 1999-08-25 2004-01-29 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
US20010036481A1 (en) 1999-08-25 2001-11-01 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
WO2001013891A2 (en) 1999-08-25 2001-03-01 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
US20010038824A1 (en) 2000-01-20 2001-11-08 Ikuo Horii Intranasal cyclic peptide formulations
WO2001060341A1 (en) 2000-02-17 2001-08-23 Rijksuniversiteit Groningen Powder formulation
US20030053960A1 (en) 2000-02-17 2003-03-20 Rijksuniversiteit Groningen Powder formulation
WO2001078696A2 (en) 2000-04-17 2001-10-25 Vectura Limited Formulations for use in inhaler devices
US20030165436A1 (en) 2000-04-17 2003-09-04 Staniforth John Nicholas Formulations for use in inhaler devices
WO2001078694A2 (en) 2000-04-17 2001-10-25 Vectura Limited Formulations for use in inhaler devices
WO2002067902A2 (en) 2001-02-23 2002-09-06 Advanced Inhalation Research, Inc. Modulation of release from dry powder formulations
WO2002080884A2 (en) 2001-03-21 2002-10-17 Boehringer Ingelheim Pharma Gmbh & Co. Kg Powder inhaler formulations
US20030019437A1 (en) 2001-07-26 2003-01-30 John Fore Ungulate game animal feed system and method
WO2003015750A1 (en) 2001-08-16 2003-02-27 Baxter International, Inc. Propellant-based microparticle formulations
WO2003072080A1 (en) 2002-02-22 2003-09-04 Advanced Inhalation Research, Inc. Inhalable formulations for sustained release
US20030232019A1 (en) 2002-02-22 2003-12-18 Advanced Inhalation Research, Inc. Inhalable formulations for sustained release
WO2003079885A2 (en) 2002-03-20 2003-10-02 Advanced Inhalation Research, Inc. Inhalable sustained therapeutic formulations
WO2003094886A2 (en) 2002-05-07 2003-11-20 Ferring Bv Desmopressin in an orodispersible dosage form
WO2005018557A2 (en) 2003-08-13 2005-03-03 Pharmacia Corporation Substituted pyridinones
EP1741703A1 (en) 2004-03-05 2007-01-10 Banyu Pharmaceutical Co., Ltd. Pyridone derivative
WO2007029847A1 (en) 2005-09-07 2007-03-15 Banyu Pharmaceutical Co., Ltd. Bicyclic aromatic substituted pyridone derivative
WO2008086404A1 (en) 2007-01-10 2008-07-17 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
"Protection for the Hydroxyl Group, Including 1,2- and 1,3-Diols", pages: 10 - 86
"Remington: The Science and Practice of Pharmacy", 2000
GOMORI, A.: "Chronic infusion of MCH causes obesity in mice", AM. J. PHYSIOL. ENDOCRINOL. METAB., vol. 284, 2002, pages E583
MCBRIAR, M. D.: "Recent advances in the discovery of melanin-concentrating hormone receptor antagonists", CURR. OPIN. DRUG DISC. & DEV., vol. 9, no. 4, 2006, pages 496
NAMING: "Indexing of Chemical Substances for Chemical Abstracts", AMERICAN CHEMICAL SOCIETY
PRAUSNITZ ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 3, 2004, pages 115
ROCKSZ, L. L.: "Biological Examination of Melanin Concentrating Hormone 1: Multi-tasking from the hypothalamus", DRUG NEWS PERSPECT, vol. 19, no. 5, 2006, pages 273
T.W.GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8268868B2 (en) 2007-01-10 2012-09-18 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles
US8273770B2 (en) 2007-07-21 2012-09-25 Albany Molecular Research, Inc. 5-pyridinone substituted indazoles
WO2019037640A1 (en) * 2017-08-22 2019-02-28 Js Innopharm (Shanghai) Ltd Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
US11242334B2 (en) 2017-08-22 2022-02-08 Js Innopharm (Shanghai) Ltd. Heterocyclic compounds as kinase inhibitors, compositions comprising the heterocyclic compound, and methods of use thereof
CN112876411A (en) * 2019-02-21 2021-06-01 杭州阿诺生物医药科技有限公司 Compound and application thereof in synthesis of PDL1 antagonist drug molecules

Also Published As

Publication number Publication date
CA2693377A1 (en) 2009-01-29
ATE544759T1 (en) 2012-02-15
KR20100044225A (en) 2010-04-29
AU2008279321B2 (en) 2013-08-01
AU2008279321A1 (en) 2009-01-29
US8273770B2 (en) 2012-09-25
EP2176251A2 (en) 2010-04-21
BRPI0814772A2 (en) 2015-03-03
US20090082359A1 (en) 2009-03-26
EP2176251B1 (en) 2012-02-08
NZ582586A (en) 2011-12-22
JP2010534248A (en) 2010-11-04
CN101861311A (en) 2010-10-13
HK1142608A1 (en) 2010-12-10
WO2009015037A3 (en) 2009-04-30
ES2382982T3 (en) 2012-06-15

Similar Documents

Publication Publication Date Title
EP2176251B1 (en) 5-pyridinone substituted indazoles and pharmaceutical compositions thereof
KR101614723B1 (en) (1-azinone) -substituted pyridoindoles as mch antagonists
US8268868B2 (en) 5-pyridinone substituted indazoles
AU2008204915B2 (en) 5-furopyridinone substituted indazoles

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200880107571.8

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 582586

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2693377

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 203358

Country of ref document: IL

Ref document number: MX/A/2010/000702

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2010518311

Country of ref document: JP

Ref document number: 373/CHENP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2008279321

Country of ref document: AU

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2008796320

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2008279321

Country of ref document: AU

Date of ref document: 20080718

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20107003768

Country of ref document: KR

Kind code of ref document: A

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08796320

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: PI0814772

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100121